Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(5): e0196954, 2018.
Article in English | MEDLINE | ID: mdl-29723289

ABSTRACT

Monoamines and neuropeptides often modulate the same behavior, but monoaminergic-peptidergic crosstalk remains poorly understood. In Caenorhabditis elegans, the adrenergic-like ligands, tyramine (TA) and octopamine (OA) require distinct subsets of neuropeptides in the two ASI sensory neurons to inhibit nociception. TA selectively increases the release of ASI neuropeptides encoded by nlp-14 or nlp-18 from either synaptic/perisynaptic regions of ASI axons or the ASI soma, respectively, and OA selectively increases the release of ASI neuropeptides encoded by nlp-9 asymmetrically, from only the synaptic/perisynaptic region of the right ASI axon. The predicted amino acid preprosequences of genes encoding either TA- or OA-dependent neuropeptides differed markedly. However, these distinct preprosequences were not sufficient to confer monoamine-specificity and additional N-terminal peptide-encoding sequence was required. Collectively, our results demonstrate that TA and OA specifically and differentially modulate the release of distinct subsets of neuropeptides from different subcellular sites within the ASIs, highlighting the complexity of monoaminergic/peptidergic modulation, even in animals with a relatively simple nervous system.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/drug effects , Neuropeptides/metabolism , Nociception/drug effects , Octopamine/pharmacology , Sensory Receptor Cells/drug effects , Tyramine/pharmacology , 1-Octanol , Amino Acid Sequence , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation , Neuropeptides/biosynthesis , Neuropeptides/genetics , Nociception/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Synapses/drug effects , Synapses/physiology
2.
Sci Rep ; 7: 45084, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338017

ABSTRACT

Among the 20 subfamilies of protein receptor tyrosine kinases (RTKs), Eph receptors are unique in possessing a sterile alpha motif (SAM domain) at their C-terminal ends. However, the functions of SAM domains in Eph receptors remain elusive. Here we report on a combined cell biology and quantitative fluorescence study to investigate the role of the SAM domain in EphA2 function. We observed elevated tyrosine autophosphorylation levels upon deletion of the EphA2 SAM domain (EphA2ΔS) in DU145 and PC3 prostate cancer cells and a skin tumor cell line derived from EphA1/A2 knockout mice. These results suggest that SAM domain deletion induced constitutive activation of EphA2 kinase activity. In order to explain these effects, we applied fluorescence correlation spectroscopy to investigate the lateral molecular organization of EphA2. Our results indicate that SAM domain deletion (EphA2ΔS-GFP) increases oligomerization compared to the full length receptor (EphA2FL-GFP). Stimulation with ephrinA1, a ligand for EphA2, induced further oligomerization and activation of EphA2FL-GFP. The SAM domain deletion mutant, EphA2ΔS-GFP, also underwent further oligomerization upon ephrinA1 stimulation, but the oligomers were larger than those observed for EphA2FL-GFP. Based on these results, we conclude that the EphA2 SAM domain inhibits kinase activity by reducing receptor oligomerization.


Subject(s)
Ephrin-A2/chemistry , Sterile Alpha Motif , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Ephrin-A1/chemistry , Ephrin-A1/metabolism , Ephrin-A2/metabolism , Humans , Mice , Phosphorylation , Protein Binding , Protein Multimerization , Protein Processing, Post-Translational , Receptor, EphA2
3.
J Neurosci ; 36(20): 5498-508, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194330

ABSTRACT

UNLABELLED: The ability to detect noxious stimuli, process the nociceptive signal, and elicit an appropriate behavioral response is essential for survival. In Caenorhabditis elegans, opioid receptor agonists, such as morphine, mimic serotonin, and suppress the overall withdrawal from noxious stimuli through a pathway requiring the opioid-like receptor, NPR-17. This serotonin- or morphine-dependent modulation can be rescued in npr-17-null animals by the expression of npr-17 or a human κ opioid receptor in the two ASI sensory neurons, with ASI opioid signaling selectively inhibiting ASI neuropeptide release. Serotonergic modulation requires peptides encoded by both nlp-3 and nlp-24, and either nlp-3 or nlp-24 overexpression mimics morphine and suppresses withdrawal. Peptides encoded by nlp-3 act differentially, with only NLP-3.3 mimicking morphine, whereas other nlp-3 peptides antagonize NLP-3.3 modulation. Together, these results demonstrate that opiates modulate nociception in Caenorhabditis elegans through a complex monoaminergic/peptidergic cascade, and suggest that this model may be useful for dissecting opiate signaling in mammals. SIGNIFICANCE STATEMENT: Opiates are used extensively to treat chronic pain. In Caenorhabditis elegans, opioid receptor agonists suppress the overall withdrawal from noxious chemical stimuli through a pathway requiring an opioid-like receptor and two distinct neuropeptide-encoding genes, with individual peptides from the same gene functioning antagonistically to modulate nociception. Endogenous opioid signaling functions as part of a complex, monoaminergic/peptidergic signaling cascade and appears to selectively inhibit neuropeptide release, mediated by a α-adrenergic-like receptor, from two sensory neurons. Importantly, receptor null animals can be rescued by the expression of the human κ opioid receptor, and injection of human opioid receptor ligands mimics exogenous opiates, highlighting the utility of this model for dissecting opiate signaling in mammals.


Subject(s)
Biogenic Monoamines/metabolism , Caenorhabditis elegans/metabolism , Neuropeptides/metabolism , Nociception , Opiate Alkaloids/pharmacology , Receptors, Opioid/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction
4.
Neuron ; 90(2): 235-44, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27041501

ABSTRACT

Thermosensation is critical for optimal regulation of physiology and behavior. C. elegans acclimates to its cultivation temperature (Tc) and exhibits thermosensitive behaviors at temperatures relative to Tc. These behaviors are mediated primarily by the AFD sensory neurons, which are extraordinarily thermosensitive and respond to thermal fluctuations at temperatures above a Tc-determined threshold. Although cGMP signaling is necessary for thermotransduction, the thermosensors in AFD are unknown. We show that AFD-specific receptor guanylyl cyclases (rGCs) are instructive for thermosensation. In addition to being necessary for thermotransduction, ectopic expression of these rGCs confers highly temperature-dependent responses onto diverse cell types. We find that the temperature response threshold is determined by the rGC and cellular context, and that multiple domains contribute to their thermosensory properties. Identification of thermosensory rGCs in C. elegans provides insight into mechanisms of thermosensation and thermal acclimation and suggests that rGCs may represent a new family of molecular thermosensors.


Subject(s)
Caenorhabditis elegans/enzymology , Caenorhabditis elegans/physiology , Receptors, Guanylate Cyclase-Coupled/physiology , Sensory Receptor Cells/physiology , Thermosensing/physiology , Animals , Animals, Genetically Modified , Muscle Cells/metabolism , Muscle Cells/physiology , Mutation , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Guanylate Cyclase-Coupled/metabolism , Sensory Receptor Cells/metabolism , Temperature , Thermosensing/genetics
5.
PLoS Pathog ; 11(4): e1004794, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25928899

ABSTRACT

Monoamines, such as 5-HT and tyramine (TA), paralyze both free-living and parasitic nematodes when applied exogenously and serotonergic agonists have been used to clear Haemonchus contortus infections in vivo. Since nematode cell lines are not available and animal screening options are limited, we have developed a screening platform to identify monoamine receptor agonists. Key receptors were expressed heterologously in chimeric, genetically-engineered Caenorhabditis elegans, at sites likely to yield robust phenotypes upon agonist stimulation. This approach potentially preserves the unique pharmacologies of the receptors, while including nematode-specific accessory proteins and the nematode cuticle. Importantly, the sensitivity of monoamine-dependent paralysis could be increased dramatically by hypotonic incubation or the use of bus mutants with increased cuticular permeabilities. We have demonstrated that the monoamine-dependent inhibition of key interneurons, cholinergic motor neurons or body wall muscle inhibited locomotion and caused paralysis. Specifically, 5-HT paralyzed C. elegans 5-HT receptor null animals expressing either nematode, insect or human orthologues of a key Gαo-coupled 5-HT1-like receptor in the cholinergic motor neurons. Importantly, 8-OH-DPAT and PAPP, 5-HT receptor agonists, differentially paralyzed the transgenic animals, with 8-OH-DPAT paralyzing mutant animals expressing the human receptor at concentrations well below those affecting its C. elegans or insect orthologues. Similarly, 5-HT and TA paralyzed C. elegans 5-HT or TA receptor null animals, respectively, expressing either C. elegans or H. contortus 5-HT or TA-gated Cl- channels in either C. elegans cholinergic motor neurons or body wall muscles. Together, these data suggest that this heterologous, ectopic expression screening approach will be useful for the identification of agonists for key monoamine receptors from parasites and could have broad application for the identification of ligands for a host of potential anthelmintic targets.


Subject(s)
Animals, Genetically Modified/metabolism , Anthelmintics/pharmacology , Behavior, Animal/drug effects , Caenorhabditis elegans/drug effects , Chloride Channel Agonists/pharmacology , Drug Discovery/methods , Serotonin 5-HT1 Receptor Agonists/pharmacology , Animals , Animals, Genetically Modified/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/agonists , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Haemonchus , Helminth Proteins/agonists , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Hypotonic Solutions/toxicity , Interneurons/drug effects , Interneurons/metabolism , Motor Activity/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Biogenic Amine/agonists , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Curr Opin Neurobiol ; 29: 17-24, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24811318

ABSTRACT

Caenorhabditis elegans navigates sensory landscapes by integrating inputs from 14 pairs of polymodal sensory neurons. Sensory neurons interact synaptically and through gap junction networks and are modulated by complex local/humoral, nutritionally dependent, monoaminergic and peptidergic signaling cascades that dynamically reconfigure individual sensory-mediated locomotory circuits. Monoaminergic/peptidergic signaling modifies the sensory signal by providing, first, feedback loops between sensory neurons and postsynaptic partners to fine tune inputs, second, crosstalk between sensory neurons to integrate responses and third, local/humoral extrasynaptic signals to facilitate broader, long term system-wide modulation. Overall, these observations highlight the differences between an anatomical wiring diagram and 'functional connectomes' that are essential to generate the alternative circuit configurations required to choose different behavioral outcomes in the face of changing environmental inputs.


Subject(s)
Nerve Net/physiology , Sensation/physiology , Sensory Receptor Cells/physiology , Synaptic Transmission/physiology , Animals , Biogenic Monoamines/metabolism , Caenorhabditis elegans , Feedback, Sensory/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction
7.
J Neurosci ; 33(35): 14107-16, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986246

ABSTRACT

Monoamines and neuropeptides interact to modulate most behaviors. To better understand these interactions, we have defined the roles of tyramine (TA), octopamine, and neuropeptides in the inhibition of aversive behavior in Caenorhabditis elegans. TA abolishes the serotonergic sensitization of aversive behavior mediated by the two nociceptive ASH sensory neurons and requires the expression of the adrenergic-like, Gαq-coupled, TA receptor TYRA-3 on inhibitory monoaminergic and peptidergic neurons. For example, TA inhibition requires Gαq and Gαs signaling in the peptidergic ASI sensory neurons, with an array of ASI neuropeptides activating neuropeptide receptors on additional neurons involved in locomotory decision-making. The ASI neuropeptides required for tyraminergic inhibition are distinct from those required for octopaminergic inhibition, suggesting that individual monoamines stimulate the release of different subsets of ASI neuropeptides. Together, these results demonstrate that a complex humoral mix of monoamines is focused by more local, synaptic, neuropeptide release to modulate nociception and highlight the similarities between the tyraminergic/octopaminergic inhibition of nociception in C. elegans and the noradrenergic inhibition of nociception in mammals that also involves inhibitory peptidergic signaling.


Subject(s)
Neuropeptides/metabolism , Nociception , Octopamine/pharmacology , Tyramine/pharmacology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Locomotion , Receptors, Catecholamine/antagonists & inhibitors , Receptors, Catecholamine/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Synaptic Transmission
8.
EMBO J ; 31(3): 667-78, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22124329

ABSTRACT

Pain modulation is complex, but noradrenergic signalling promotes anti-nociception, with α(2)-adrenergic agonists used clinically. To better understand the noradrenergic/peptidergic modulation of nociception, we examined the octopaminergic inhibition of aversive behaviour initiated by the Caenorhabditis elegans nociceptive ASH sensory neurons. Octopamine (OA), the invertebrate counterpart of norepinephrine, modulates sensory-mediated reversal through three α-adrenergic-like OA receptors. OCTR-1 and SER-3 antagonistically modulate ASH signalling directly, with OCTR-1 signalling mediated by Gα(o). In contrast, SER-6 inhibits aversive responses by stimulating the release of an array of 'inhibitory' neuropeptides that activate receptors on sensory neurons mediating attraction or repulsion, suggesting that peptidergic signalling may integrate multiple sensory inputs to modulate locomotory transitions. These studies highlight the complexity of octopaminergic/peptidergic interactions, the role of OA in activating global peptidergic signalling cascades and the similarities of this modulatory network to the noradrenergic inhibition of nociception in mammals, where norepinephrine suppresses chronic pain through inhibitory α(2)-adrenoreceptors on afferent nociceptors and stimulatory α(1)-receptors on inhibitory peptidergic interneurons.


Subject(s)
Avoidance Learning , Biogenic Monoamines/metabolism , Caenorhabditis elegans/physiology , Neuropeptides/metabolism , 1-Octanol/pharmacology , Animals , Animals, Genetically Modified , GTP-Binding Proteins/metabolism , Polymerase Chain Reaction , Serotonin/pharmacology , Signal Transduction , Xenopus laevis
9.
Worm ; 1(4): 202-6, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-24058849

ABSTRACT

Octopamine (OA) appears to function as the invertebrate counterpart of norepinephrine (NE) in the modulation of a number of key behaviors. In C. elegans, OA signaling is complex, mediated by at least three distinct α-adrenergic-like receptors and appears to activate more global peptidergic signaling cascades that have the potential to dramatically amplify the octopaminergic signal. These OA-dependent peptidergic signaling cascades involve an array of neuropeptides that activate receptors throughout the nervous system and have the potential to both directly and indirectly modulate locomotory decision-making. In this commentary we highlight the use of C. elegans as a model to expand our understanding of noradrenergic signaling in mammals, specifically as it relates to the role of NE in anti-nociception.

10.
Invert Neurosci ; 12(1): 53-61, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22143253

ABSTRACT

Monoamines and neuropeptides interact to modulate key behaviors in most organisms. This review is focused on the interaction between octopamine (OA) and an array of neuropeptides in the inhibition of a simple, sensory-mediated aversive behavior in the C. elegans model system and describes the role of monoamines in the activation of global peptidergic signaling cascades. OA has been often considered the invertebrate counterpart of norepinephrine, and the review also highlights the similarities between OA inhibition in C. elegans and the noradrenergic modulation of pain in higher organisms.


Subject(s)
Chronic Pain/metabolism , Disease Models, Animal , Neuropeptides/metabolism , Octopamine/metabolism , Signal Transduction/physiology , Animals , Behavior, Animal , Biogenic Monoamines/metabolism , Caenorhabditis elegans/metabolism , Chronic Pain/physiopathology , Nociception/physiology
11.
PLoS One ; 6(7): e21897, 2011.
Article in English | MEDLINE | ID: mdl-21814562

ABSTRACT

Nutritional state often modulates olfaction and in Caenorhabditis elegans food stimulates aversive responses mediated by the nociceptive ASH sensory neurons. In the present study, we have characterized the role of key serotonergic neurons that differentially modulate aversive behavior in response to changing nutritional status. The serotonergic NSM and ADF neurons play antagonistic roles in food stimulation. NSM 5-HT activates SER-5 on the ASHs and SER-1 on the RIA interneurons and stimulates aversive responses, suggesting that food-dependent serotonergic stimulation involves local changes in 5-HT levels mediated by extrasynaptic 5-HT receptors. In contrast, ADF 5-HT activates SER-1 on the octopaminergic RIC interneurons to inhibit food-stimulation, suggesting neuron-specific stimulatory and inhibitory roles for SER-1 signaling. Both the NSMs and ADFs express INS-1, an insulin-like peptide, that appears to cell autonomously inhibit serotonergic signaling. Food also modulates directional decisions after reversal is complete, through the same serotonergic neurons and receptors involved in the initiation of reversal, and the decision to continue forward or change direction after reversal is dictated entirely by nutritional state. These results highlight the complexity of the "food signal" and serotonergic signaling in the modulation of sensory-mediated aversive behaviors.


Subject(s)
Behavior, Animal/physiology , Diet , Interneurons/metabolism , Nociceptors/metabolism , Sensory Receptor Cells/metabolism , Serotonergic Neurons/metabolism , Serotonin/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/physiology , Insulin/metabolism , Signal Transduction
12.
Genetics ; 186(3): 929-41, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739712

ABSTRACT

Fluoxetine is one of the most commonly prescribed medications for many behavioral and neurological disorders. Fluoxetine acts primarily as an inhibitor of the serotonin reuptake transporter (SERT) to block the removal of serotonin from the synaptic cleft, thereby enhancing serotonin signals. While the effects of fluoxetine on behavior are firmly established, debate is ongoing whether inhibition of serotonin reuptake is a sufficient explanation for its therapeutic action. Here, we provide evidence of two additional aspects of fluoxetine action through genetic analyses in Caenorhabditis elegans. We show that fluoxetine treatment and null mutation in the sole SERT gene mod-5 eliminate serotonin in specific neurons. These neurons do not synthesize serotonin but import extracellular serotonin via MOD-5/SERT. Furthermore, we show that fluoxetine acts independently of MOD-5/SERT to regulate discrete properties of acetylcholine (Ach), gamma-aminobutyric acid (GABA), and glutamate neurotransmission in the locomotory circuit. We identified that two G-protein-coupled 5-HT receptors, SER-7 and SER-5, antagonistically regulate the effects of fluoxetine and that fluoxetine binds to SER-7. Epistatic analyses suggest that SER-7 and SER-5 act upstream of AMPA receptor GLR-1 signaling. Our work provides genetic evidence that fluoxetine may influence neuronal functions and behavior by directly targeting serotonin receptors.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Fluoxetine/pharmacology , Synaptic Transmission/drug effects , Acetylcholine/metabolism , Animals , Behavior, Animal/drug effects , Biological Assay , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Fluoxetine/metabolism , Glutamic Acid/metabolism , Muscle Relaxation/drug effects , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Serotonin/metabolism , Serotonin/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction/drug effects , gamma-Aminobutyric Acid/metabolism
13.
J Neurosci ; 30(23): 7889-99, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20534837

ABSTRACT

Monoamines and neuropeptides interact to modulate behavioral plasticity in both vertebrates and invertebrates. In Caenorhabditis elegans behavioral state or "mood" is dependent on food availability and is translated by both monoaminergic and peptidergic signaling in the fine-tuning of most behaviors. In the present study, we have examined the interaction of monoamines and peptides on C. elegans aversive behavior mediated by a pair of polymodal, nociceptive, ASH sensory neurons. Food or serotonin sensitize the ASHs and stimulate aversive responses through a pathway requiring the release of nlp-3-encoded neuropeptides from the ASHs. Peptides encoded by nlp-3 appear to stimulate ASH-mediated aversive behavior through the neuropeptide receptor-17 (NPR-17) receptor. nlp-3- and npr-17-null animals exhibit identical phenotypes and animals overexpressing either nlp-3 or npr-17 exhibit elevated aversive responses off food that are absent when nlp-3 or npr-17 are overexpressed in npr-17- or nlp-3-null animals, respectively. ASH-mediated aversive responses are increased by activating either Galpha(q) or Galpha(s) in the ASHs, with Galpha(s) signaling specifically stimulating the release of nlp-3-encoded peptides. In contrast, octopamine appears to inhibit 5-HT stimulation by activating Galpha(o) signaling in the ASHs that, in turn, inhibits both Galpha(s) and Galpha(q) signaling and the release of nlp-3-encoded peptides. These results demonstrate that serotonin and octopamine reversibly modulate the activity of the ASHs, and highlight the utility of the C. elegans model for defining interactions between monoamines and peptides in individual neurons of complex sensory-mediated circuits.


Subject(s)
Biogenic Monoamines/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Neuropeptides/metabolism , Nociceptors/metabolism , Signal Transduction/drug effects , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Biogenic Monoamines/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Nociceptors/drug effects , Octanols/adverse effects , Octopamine/pharmacology , Serotonin/pharmacology
14.
J Neurosci ; 29(5): 1446-56, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19193891

ABSTRACT

Serotonin modulates behavioral plasticity in both vertebrates and invertebrates and in Caenorhabditis elegans regulates key behaviors, including locomotion, aversive learning and olfaction through at least four different 5-HT receptors. In the present study, we examined the serotonergic stimulation of aversive responses to dilute octanol in animals containing null alleles of these 5-HT receptors. Both ser-1 and mod-1 null animals failed to increase sensitivity to dilute octanol on food/5-HT, in contrast to wild-type, ser-4 or ser-7 null animals. 5-HT sensitivity was restored by the expression of MOD-1 and SER-1 in the AIB or potentially the AIY, and RIA interneurons of mod-1 and ser-1 null animals, respectively. Because none of these 5-HT receptors appear to be expressed in the ASH sensory neurons mediating octanol sensitivity, we identified a 5-HT(6)-like receptor, F16D3.7(SER-5), that was required for food/5-HT-dependent increases in octanol sensitivity. ser-5 null animals failed to increase octanol sensitivity in the presence of food/5-HT and sensitivity could be restored by expression of SER-5 in the ASHs. Similarly, the RNAi knockdown of ser-5 expression in the ASHs of wild-type animals also abolished 5-HT-dependent increases in octanol sensitivity, suggesting that SER-5 modulates the octanol responsiveness of the ASHs directly. Together, these results suggest that multiple amine receptors, functioning at different levels within the locomotory circuit, are each essential for the serotonergic modulation of ASH-mediated aversive responses.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Chemoreceptor Cells/physiology , Motor Activity/physiology , Nerve Net/physiology , Receptors, Serotonin/physiology , Serotonin/physiology , 1-Octanol/pharmacology , Amino Acid Sequence , Animals , COS Cells , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/physiology , Chlorocebus aethiops , Gene Knockdown Techniques/methods , Interneurons/physiology , Molecular Sequence Data , Motor Activity/genetics , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT2/genetics , Receptors, Serotonin, 5-HT2/physiology , Serotonin/deficiency , Serotonin/genetics , Signal Transduction/physiology
15.
Genetics ; 181(1): 153-63, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001289

ABSTRACT

Serotonin (5-HT) regulates key processes in both vertebrates and invertebrates. Previously, four 5-HT receptors that contributed to the 5-HT modulation of egg laying were identified in Caenorhabditis elegans. Therefore, to assess potential receptor interactions, we generated animals containing combinations of null alleles for each receptor, especially animals expressing only individual 5-HT receptors. 5-HT-stimulated egg laying and egg retention correlated well with different combinations of predicted excitatory and inhibitory serotonergic inputs. For example, 5-HT did not stimulate egg laying in ser-1, ser-7, or ser-7 ser-1 null animals, and ser-7 ser-1 animals retained more eggs than wild-type animals. In contrast, 5-HT-stimulated egg laying in ser-4;mod-1 animals was greater than in wild-type animals, and ser-4;mod-1 animals retained fewer eggs than wild-type animals. Surprisingly, ser-4;mod-1;ser-7 ser-1 animals retained the same number of eggs as wild-type animals and exhibited significant 5-HT-stimulated egg laying that was dependent on a previously uncharacterized receptor, SER-5. 5-HT-stimulated egg laying was absent in ser-5;ser-4;mod-1;ser-7 ser-1 animals, and these animals retained more eggs than either wild-type or ser-4;mod-1;ser-7 ser-1 animals. The 5-HT sensitivity of egg laying could be restored by ser-5 muscle expression. Together, these results highlight the dual excitatory/inhibitory serotonergic inputs that combine to modulate egg laying.


Subject(s)
Caenorhabditis elegans/physiology , Oviposition/physiology , Serotonin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Female , Locomotion/drug effects , Models, Biological , Molecular Sequence Data , Muscles/drug effects , Muscles/metabolism , Mutation/genetics , Oviposition/drug effects , Phylogeny , Receptors, Serotonin/chemistry , Serotonin/pharmacology , Signal Transduction/drug effects
16.
J Neurosci ; 27(49): 13402-12, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-18057198

ABSTRACT

Biogenic amines modulate key behaviors in both vertebrates and invertebrates. In Caenorhabditis elegans, tyramine (TA) and octopamine (OA) inhibit aversive responses to 100%, but not dilute (30%) octanol. TA and OA also abolish food- and serotonin-dependent increases in responses to dilute octanol in wild-type but not tyra-3(ok325) and f14d12.6(ok371) null animals, respectively, suggesting that TA and OA modulated responses to dilute octanol are mediated by separate, previously uncharacterized, G-protein-coupled receptors. TA and OA are high-affinity ligands for TYRA-3 and F14D12.6, respectively, based on their pharmacological characterization after heterologous expression. f14d12.6::gfp is expressed in the ASHs, the neurons responsible for sensitivity to dilute octanol, and the sra-6-dependent expression of F14D12.6 in the ASHs is sufficient to rescue OA sensitivity in f14d12.6(ok371) null animals. In contrast, tyra-3::gfp appears not to be expressed in the ASHs, but instead in other neurons, including the dopaminergic CEP/ADEs. However, although dopamine (DA) also inhibits 5-HT-dependent responses to dilute octanol, TA still inhibits in dop-2; dop-1; dop-3 animals that do not respond to DA and cat-2(tm346) and Pdat-1::ICE animals that lack significant dopaminergic signaling, suggesting that DA is not an intermediate in TA inhibition. Finally, responses to TA and OA selectively desensitize after preexposure to the amines. Our data suggest that although tyraminergic and octopaminergic signaling yield identical phenotypes in these olfactory assays, they act independently through distinct receptors to modulate the ASH-mediated locomotory circuit and that C. elegans is a useful model to study the aminergic modulation of sensory-mediated locomotory behaviors.


Subject(s)
Behavior, Animal/physiology , Caenorhabditis elegans Proteins/physiology , Octopamine/physiology , Receptors, Biogenic Amine/physiology , Serotonin/physiology , Tyramine/physiology , Animals , CHO Cells , COS Cells , Caenorhabditis elegans , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/antagonists & inhibitors , Chlorocebus aethiops , Cricetinae , Cricetulus , Humans , Mice , NIH 3T3 Cells , Octopamine/pharmacology , Phylogeny , Receptors, Biogenic Amine/agonists , Receptors, Biogenic Amine/antagonists & inhibitors , Serotonin/pharmacology , Tyramine/pharmacology
17.
Dev Biol ; 298(2): 379-91, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16890216

ABSTRACT

Serotonin (5-HT) stimulation of egg-laying in Caenorhabditis elegans is abolished in ser-1 (ok345) animals and is rescued by ser-1 expression in vulval muscle. A PDZ binding motif (ETFL) at the SER-1 C-terminus is not essential for rescue, but facilitates SER-1 signaling. SER-1 binds specifically to PDZ domain 10 of the multi-PDZ domain protein, MPZ-1, based on GST pulldown and co-immunoprecipitation. mpz-1 is expressed in about 60 neurons and body wall and vulval muscles. In neurons, GFP-tagged MPZ-1 is punctate and colocalizes with the synaptic marker, synaptobrevin. The expression patterns of ser-1 and mpz-1 overlap in 3 pairs of neurons and vulval muscle. In addition, MPZ-1 also interacts with other GPCRs with acidic amino acids in the -3 position of their PDZ binding motifs. mpz-1 RNAi reduces 5-HT stimulated egg-laying in wild type animals and in ser-1 mutants rescued by muscle expression of SER-1. In contrast, mpz-1 RNAi has no effect on 5-HT stimulated egg-laying in ser-1 mutants rescued by expression of a truncated SER-1 that lacks the C-terminal PDZ binding motif. The overexpression of MPZ-1 PDZ domain 10 also inhibits 5-HT stimulated egg-laying. These studies suggest that the SER-1/MPZ-1 interaction facilitates SER-1 mediated signaling.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Muscles/metabolism , Receptors, Serotonin, 5-HT2/physiology , Serotonin/pharmacology , Animals , Base Sequence , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Eggs , Female , Molecular Sequence Data , Muscles/physiology , Neurons/metabolism , Protein Structure, Tertiary , RNA Interference , Receptors, G-Protein-Coupled , Receptors, Serotonin, 5-HT2/genetics , Receptors, Serotonin, 5-HT2/metabolism , Sequence Homology, Nucleic Acid , Signal Transduction , Vulva/metabolism , Vulva/physiology
18.
Genetics ; 172(1): 159-69, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16204223

ABSTRACT

Serotonin (5-HT) stimulates both pharyngeal pumping and egg laying in Caenorhabditis elegans. Four distinct 5-HT receptors have been partially characterized, but little is known about their function in vivo. SER-7 exhibits most sequence identity to the mammalian 5-HT7 receptors and couples to a stimulation of adenyl cyclase when expressed in COS-7 cells. However, many 5-HT7-specific agonists have low affinity for SER-7. 5-HT fails to stimulate pharyngeal pumping and the firing of the MC motorneurons in animals containing the putative ser-7(tm1325) and ser-7(tm1728) null alleles. In addition, although pumping on bacteria is upregulated in ser-7(tm1325) animals, pumping is more irregular. A similar failure to maintain "fast pumping" on bacteria also was observed in ser-1(ok345) and tph-1(mg280) animals that contain putative null alleles of a 5-HT2-like receptor and tryptophan hydroxylase, respectively, suggesting that serotonergic signaling, although not essential for the upregulation of pumping on bacteria, "fine tunes" the process. 5-HT also fails to stimulate egg laying in ser-7(tm1325), ser-1(ok345), and ser-7(tm1325) ser-1(ok345) animals, but only the ser-7 ser-1 double mutants exhibit an Egl phenotype. All of the SER-7 mutant phenotypes are rescued by the expression of full-length ser-7gfp translational fusions. ser-7gfp is expressed in several pharyngeal neurons, including the MC, M2, M3, M4, and M5, and in vulval muscle. Interestingly, 5-HT inhibits egg laying and pharyngeal pumping in ser-7 null mutants and the 5-HT inhibition of egg laying, but not pumping, is abolished in ser-7(tm1325);ser-4(ok512) double mutants. Taken together, these results suggest that SER-7 is essential for the 5-HT stimulation of both egg laying and pharyngeal pumping, but that other signaling pathways can probably fulfill similar roles in vivo.


Subject(s)
Caenorhabditis elegans/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Oviposition/physiology , Pharynx/metabolism , Serotonin/pharmacology , Adenylyl Cyclases/metabolism , Animals , Behavior, Animal , COS Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Chlorocebus aethiops , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Ligands , Motor Neurons/metabolism , Muscles/physiology , Oviposition/drug effects , Pharynx/drug effects , Phenotype , Receptors, Serotonin, 5-HT2/chemistry , Receptors, Serotonin, 5-HT2/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Signal Transduction , Tryptophan Hydroxylase/chemistry , Tryptophan Hydroxylase/metabolism , Vulva/physiology
19.
J Neurochem ; 94(1): 181-91, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15953361

ABSTRACT

Tyramine appears to regulate key processes in nematodes, such as pharyngeal pumping, and more complex behaviors, such as foraging. Recently, a Caenorhabditis elegans tyramine receptor, SER-2, was identified that is involved in the TA-dependent regulation of these processes. In the present study, we have identified a second C. elegans gene, tyra-2 (F01E11.5) that encodes a tyramine receptor. This is the first identification of multiple tyramine receptor genes in any invertebrate. Membranes from COS-7 cells expressing TYRA-2 bind [(3)H]tyramine with high affinity with a K(d) of 20 +/- 5 nM. Other physiologically relevant biogenic amines, such as octopamine and dopamine, inhibit [(3)H]tyramine binding with much lower affinity (K(i)s of 1.55 +/- 0.5 and 1.78 +/- 0.6 microM, respectively), supporting the identification of TYRA-2 as a tyramine receptor. Indeed, tyramine also dramatically increases GTPgammaS binding to membranes from cells expressing TYRA-2 (EC(50) of 50 +/- 13 nM) and the TA-dependent GTPgammaS binding is PTX-sensitive suggesting that TYRA-2 may couple to Galpha(i/o). Based on fluorescence from tyra::gfp fusion constructs, TYRA-2 expression appears to be exclusively neuronal in the MC and NSM pharyngeal neurons, the AS family of amphid neurons and neurons in the nerve ring, body and tail. Taken together, these results suggest that TYRA-2 encodes a second Galpha(i/o)-coupled tyramine receptor and suggests that TA-dependent neuromodulation may be mediated by multiple receptors and more complex than previously appreciated.


Subject(s)
Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans/metabolism , Motor Neurons/metabolism , Pharynx/metabolism , Receptors, Biogenic Amine/biosynthesis , Tyramine/metabolism , Amino Acid Sequence , Animals , COS Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Molecular Sequence Data , Motor Neurons/physiology , Pharynx/cytology , Pharynx/physiology , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/physiology
20.
J Neurochem ; 91(5): 1104-15, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15569254

ABSTRACT

Octopamine regulates essential processes in nematodes; however, little is known about the physiological role of its precursor, tyramine. In the present study, we have characterized alternatively spliced Caenorhabditis elegans tyramine receptor isoforms (SER-2 and SER-2A) that differ by 23 amino acids within the mid-region of the third intracellular loop. Membranes prepared from cells expressing either SER-2 or SER-2A bind [3H]lysergic acid diethylamide (LSD) in the low nanomolar range and exhibit highest affinity for tyramine. Similarly, both isoforms exhibit nearly identical Ki values for a number of antagonists. In contrast, SER-2A exhibits a significantly lower affinity than SER-2 for other physiologically relevant biogenic amines, including octopamine. Pertussis toxin treatment reduces affinity for both tyramine and octopamine, especially for octopamine in membranes from cells expressing SER-2, suggesting that the conformation of the mid-region of the third intracellular loop is dictated by G-protein interactions and is responsible for the differential tyramine/octopamine affinities of the two isoforms. Tyramine reduces forskolin-stimulated cAMP levels in HEK293 cells expressing either isoform with nearly identical IC50 values. Tyramine, but not octopamine, also elevates Ca2+ levels in cells expressing SER-2 and to a lesser extent SER-2A. Most importantly, ser-2 null mutants (pk1357) fail to suppress head movements while reversing in response to nose-touch, suggesting a role for SER-2 in the regulation of foraging behavior, and fail to respond to tyramine in assays measuring serotonin-dependent pharyngeal pumping. These are the first reported functions for SER-2. These results suggest that C. elegans contains tyramine receptors, that individual SER-2 isoforms may differ significantly in their sensitivity to other physiologically relevant biogenic amines, such as octopamine (OA), and that tyraminergic signaling may be important in the regulation of key processes in nematodes.


Subject(s)
Feeding Behavior/physiology , Pharynx/physiology , Protein Isoforms/physiology , Receptors, Biogenic Amine/physiology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Agonists/pharmacology , Amino Acid Sequence , Animals , Animals, Genetically Modified/physiology , Behavior, Animal , Biogenic Monoamines/pharmacokinetics , Caenorhabditis elegans/physiology , Calcium/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Cloning, Molecular/methods , Cyclic AMP/metabolism , DNA, Recombinant , Diagnostic Imaging/methods , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Embryo, Nonmammalian , Extracellular Space/metabolism , Gene Expression/physiology , Green Fluorescent Proteins/metabolism , Humans , Lysergic Acid Diethylamide/pharmacokinetics , Models, Molecular , Nose/drug effects , Nose/physiology , Octopamine/pharmacology , Pertussis Toxin/pharmacology , Phenotype , Phosphatidylinositols/metabolism , RNA, Messenger/biosynthesis , Radioligand Assay/methods , Receptors, Biogenic Amine/chemistry , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Serotonin/pharmacology , Time Factors , Transfection , Tritium/pharmacokinetics , Tyramine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...