Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 184: 108415, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33275959

ABSTRACT

Several studies have shown that ethanol (EtOH) can enhance the activity of GABAergic synapses via presynaptic mechanisms, including in hippocampal CA1 neurons. The serotonin type 3 receptor (5-HT3-R) has been implicated in the neural actions of ethanol (EtOH) and in modulation of GABA release from presynaptic terminals. In the present study, we investigated EtOH modulation of GABA release induced by 5-HT3-R activation using the mechanically isolated neuron/bouton preparation from the rat CA1 hippocampal subregion. EtOH application before and during exposure to the selective 5-HT3 receptor agonist, m-chlorophenylbiguanide (mCPBG) potentiated the mCPBG-induced increases in the peak frequency and charge transfer of spontaneous GABAergic inhibitory postsynaptic currents. Interestingly, the potentiation was maintained even after EtOH was removed from the preparation. A protein kinase A inhibitor reduced the magnitude of EtOH potentiation. Fluorescent Ca2+ imaging showed that Ca2+ transients in the presynaptic terminals increased during EtOH exposure. These findings indicate that EtOH produces long-lasting potentiation of 5-HT3-induced GABA release by modulating calcium levels, via a process involving cAMP-mediated signaling in presynaptic terminals.


Subject(s)
CA1 Region, Hippocampal/metabolism , Ethanol/administration & dosage , Neurons/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Biguanides/administration & dosage , CA1 Region, Hippocampal/drug effects , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Female , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/administration & dosage , Synapses/drug effects
2.
Front Neurosci ; 14: 615362, 2020.
Article in English | MEDLINE | ID: mdl-33424545

ABSTRACT

Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.

3.
Sci Rep ; 8(1): 17379, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478340

ABSTRACT

Activation of short-chain free fatty acid receptors 3 (FFAR3) has been suggested to promote sympathetic outflow in postganglionic sympathetic neurons or hamper it by a negative coupling to N-type calcium (CaV2.2) channels. Heterogeneity of FFAR3 expression in sympathetic neurons, however, renders single neurons studies extremely time-consuming in wild-type mice. Previous studies demonstrated large variability of the degree of CaV2.2 channel inhibition by FFAR3 in a global population of rat sympathetic neurons. Therefore, we focused on a small subpopulation of mouse sympathetic neurons using an FFAR3 antibody and an Ffar3 reporter mouse to perform immunofluorescent and electrophysiological studies. Whole-cell patch-clamp recordings of identified FFAR3-expressing neurons from reporter mice revealed a 2.5-fold decrease in the CaV2.2-FFAR3 inhibitory coupling variability and 1.5-fold increase in the mean ICa2+ inhibition, when compared with unlabeled neurons from wild-type mice. Further, we found that the ablation of Ffar3 gene expression in two knockout mouse models led to a complete loss-of-function. Subpopulations of sympathetic neurons are associated with discrete functional pathways. However, little is known about the neural pathways of the FFAR3-expressing subpopulation. Our data indicate that FFAR3 is expressed primarily in neurons with a vasoconstrictor phenotype. Thus, fine-tuning of chemically-coded neurotransmitters may accomplish an adequate outcome.


Subject(s)
Adrenergic Fibers/metabolism , Calcium Channels, N-Type/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Female , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neurons , Patch-Clamp Techniques/methods , Signal Transduction/physiology
4.
Cold Spring Harb Protoc ; 2016(5)2016 05 02.
Article in English | MEDLINE | ID: mdl-27140920

ABSTRACT

Sympathetic neurons isolated from adult rat superior cervical ganglia (SCG) are a well-established model to study G-protein modulation of voltage-gated Ca(2+) channels (VGCCs). SCG neurons can be easily dissociated and are amendable to heterologous expression of genes, including genetic tools to study G-protein signaling pathways, within a time frame to maintain good spatial voltage-clamp control of membrane potential during electrophysiological recordings (8-36 h postdissociation). This protocol focuses on examining G-protein modulation of VGCCs; however, the procedures and experimental setup for acute application of agonists can be applied to study modulation of other ion channels (e.g., M-current, G-protein-coupled inwardly rectifying K(+) channels). We also discuss some common sources of artifacts that can arise during acute drug application onto dissociated neurons, which can mislead interpretation of results.


Subject(s)
Calcium Channels, N-Type/metabolism , GTP-Binding Proteins/metabolism , Neurons/physiology , Patch-Clamp Techniques/methods , Superior Cervical Ganglion/cytology , Animals , Calcium Channels, N-Type/genetics , GTP-Binding Proteins/genetics , Rats
5.
Cold Spring Harb Protoc ; 2016(5)2016 05 02.
Article in English | MEDLINE | ID: mdl-27140924

ABSTRACT

G-protein-coupled receptor modulation of voltage-gated ion channels is a common means of fine-tuning the response of channels to changes in membrane potential. Such modulation impacts physiological processes such as synaptic transmission, and hence therapeutic strategies often directly or indirectly target these pathways. As an exemplar of channel modulation, we examine strategies for investigating G-protein modulation of CaV2.2 or N-type voltage-gated Ca(2+) channels. We focus on biochemical and genetic tools for defining the molecular mechanisms underlying the various forms of CaV2.2 channel modulation initiated following ligand binding to G-protein-coupled receptors.


Subject(s)
Calcium Channels, N-Type/metabolism , Cytological Techniques/methods , GTP-Binding Proteins/metabolism , Molecular Biology/methods , Receptors, G-Protein-Coupled/metabolism , Animals , Calcium Channels, N-Type/genetics , GTP-Binding Proteins/genetics , Humans , Receptors, G-Protein-Coupled/genetics
6.
Sci Rep ; 6: 25137, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27118437

ABSTRACT

Rem2 is a member of the RGK subfamily of RAS small GTPases. Rem2 inhibits high voltage activated calcium channels, is involved in synaptogenesis, and regulates dendritic morphology. Rem2 is the primary RGK protein expressed in the nervous system, but to date, the precise expression patterns of this protein are unknown. In this study, we characterized Rem2 expression in the mouse nervous system. In the CNS, Rem2 mRNA was detected in all regions examined, but was enriched in the striatum. An antibody specific for Rem2 was validated using a Rem2 knockout mouse model and used to show abundant expression in striatonigral and striatopallidal medium spiny neurons but not in several interneuron populations. In the PNS, Rem2 was abundant in a subpopulation of neurons in the trigeminal and dorsal root ganglia, but was absent in sympathetic neurons of superior cervical ganglia. Under basal conditions, Rem2 was subject to post-translational phosphorylation, likely at multiple residues. Further, Rem2 mRNA and protein expression peaked at postnatal week two, which corresponds to the period of robust neuronal maturation in rodents. This study will be useful for elucidating the functions of Rem2 in basal ganglia physiology.


Subject(s)
Basal Ganglia/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Animals , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Mice , Nervous System/metabolism , Phosphorylation , Protein Processing, Post-Translational , Trigeminal Ganglion/metabolism
7.
Sci Rep ; 5: 12880, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26260360

ABSTRACT

FFAR3 (GPR41) is a G-protein coupled receptor for which short-chain fatty acids serve as endogenous ligands. The receptor is found on gut enteroendocrine L-cells, pancreatic ß-cells, and sympathetic neurons, and is implicated in obesity, diabetes, allergic airway disease, and altered immune function. In primates, FFAR3 is segmentally duplicated resulting in GPR42, a gene currently classified as a suspected pseudogene. In this study, we sequenced FFAR3 and GPR42 open reading frames from 56 individuals and found an unexpectedly high frequency of polymorphisms contributing to several complex haplotypes. We also identified a frequent (18.8%) structural variation that results in GPR42 copy number polymorphism. Finally, sequencing revealed that 50.6% of GPR42 haplotypes differed from FFAR3 by only a single non-synonymous substitution and that the GPR42 reference sequence matched only 4.4% of the alleles. Sequencing of cDNA from human sympathetic ganglia and colon revealed processed transcripts matching the GPR42 genotype. Expression of several GPR42 haplotypes in rat sympathetic neurons revealed diverse pharmacological phenotypes that differed in potency and efficacy. Our data suggest that GPR42 be reclassified as a functioning gene and that recognition of sequence and copy number polymorphism of the FFAR3/GPR42 complex be considered during genetic and pharmacological investigation of these receptors.


Subject(s)
DNA Copy Number Variations/genetics , Pseudogenes , Receptors, G-Protein-Coupled/genetics , Animals , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Genotype , Haplotypes/genetics , Humans , Neurons/drug effects , Neurons/metabolism , Rats , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/biosynthesis
8.
Mol Pharmacol ; 88(2): 238-44, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26013543

ABSTRACT

6-Alkoxy-5-aryl-3-pyridincarboxamides, including the brain-penetrant compound 14G: [5-(4-chlorophenyl)-6-(cyclopropylmethoxy)-N-[(1R,2R)-2-hydroxy-cyclohexyl]-3-pyridinecarboxamide] and its peripherally restricted analog 14H: [5-(4-chlorophenyl)-N-[(1R,2R)-2-hydroxycyclohexyl]-6-(2-methoxyethoxy)-3-pyridinecarboxamide], have been recently introduced as selective, high-affinity antagonists of the human cannabinoid-1 receptor (hCB1R). Binding analyses revealed two orders of magnitude lower affinity of these compounds for mouse and rat versus human CB1R, whereas the affinity of rimonabant is comparable for all three CB1Rs. Modeling of ligand binding to CB1R and binding assays with native and mutant (Ile105Met) hCB1Rs indicate that the Ile105 to Met mutation in rodent CB1Rs accounts for the species-dependent affinity of 14G: and 14H: . Our work identifies Ile105 as a new pharmacophore component for developing better hCB1R antagonists and invalidates rodent models for assessing the antiobesity efficacy of 14G: and 14H: .


Subject(s)
Brain/metabolism , Cannabinoid Receptor Antagonists/chemical synthesis , Cannabinoid Receptor Antagonists/pharmacology , Niacinamide/analogs & derivatives , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/genetics , Animals , Cannabinoid Receptor Antagonists/chemistry , HEK293 Cells , Humans , Isoleucine/metabolism , Mice , Models, Molecular , Niacinamide/chemical synthesis , Niacinamide/chemistry , Niacinamide/pharmacology , Piperidines/chemistry , Pyrazoles/chemistry , Rats , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Species Specificity , Structure-Activity Relationship , X-Ray Diffraction
9.
J Neurosci ; 35(20): 8021-34, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995484

ABSTRACT

Under physiological conditions, the voltage-gated sodium channel Nav1.8 is expressed almost exclusively in primary sensory neurons. The mechanism restricting Nav1.8 expression is not entirely clear, but we have previously described a 3.7 kb fragment of the Scn10a promoter capable of recapitulating the tissue-specific expression of Nav1.8 in transfected neurons and cell lines (Puhl and Ikeda, 2008). To validate these studies in vivo, a transgenic mouse encoding EGFP under the control of this putative sensory neuron specific promoter was generated and characterized in this study. Approximately 45% of dorsal root ganglion neurons of transgenic mice were EGFP-positive (mean diameter = 26.5 µm). The majority of EGFP-positive neurons bound isolectin B4, although a small percentage (∼10%) colabeled with markers of A-fiber neurons. EGFP expression correlated well with the presence of Nav1.8 transcript (95%), Nav1.8-immunoreactivity (70%), and TTX-R INa (100%), although not all Nav1.8-expressing neurons expressed EGFP. Several cranial sensory ganglia originating from neurogenic placodes, such as the nodose ganglion, failed to express EGFP, suggesting that additional regulatory elements dictate Scn10a expression in placodal-derived sensory neurons. EGFP was also detected in discrete brain regions of transgenic mice. Quantitative PCR and Nav1.8-immunoreactivity confirmed Nav1.8 expression in the amygdala, brainstem, globus pallidus, lateral and paraventricular hypothalamus, and olfactory tubercle. TTX-R INa recorded from EGFP-positive hypothalamic neurons demonstrate the usefulness of this transgenic line to study novel roles of Nav1.8 beyond sensory neurons. Overall, Scn10a-EGFP transgenic mice recapitulate the majority of the Nav1.8 expression pattern in neural crest-derived sensory neurons.


Subject(s)
Cell Lineage , Green Fluorescent Proteins/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neural Crest/metabolism , Promoter Regions, Genetic , Animals , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , Nerve Fibers, Myelinated/metabolism , Neural Crest/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Organ Specificity , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
10.
PLoS One ; 9(7): e100694, 2014.
Article in English | MEDLINE | ID: mdl-24992013

ABSTRACT

RGK proteins, Gem, Rad, Rem1, and Rem2, are members of the Ras superfamily of small GTP-binding proteins that interact with Ca2+ channel ß subunits to modify voltage-gated Ca2+ channel function. In addition, RGK proteins affect several cellular processes such as cytoskeletal rearrangement, neuronal dendritic complexity, and synapse formation. To probe the phylogenetic origins of RGK protein-Ca2+ channel interactions, we identified potential RGK-like protein homologs in genomes for genetically diverse organisms from both the deuterostome and protostome animal superphyla. RGK-like protein homologs cloned from Danio rerio (zebrafish) and Drosophila melanogaster (fruit flies) expressed in mammalian sympathetic neurons decreased Ca2+ current density as reported for expression of mammalian RGK proteins. Sequence alignments from evolutionarily diverse organisms spanning the protostome/deuterostome divide revealed conservation of residues within the RGK G-domain involved in RGK protein--Cavß subunit interaction. In addition, the C-terminal eleven residues were highly conserved and constituted a signature sequence unique to RGK proteins but of unknown function. Taken together, these data suggest that RGK proteins, and the ability to modify Ca2+ channel function, arose from an ancestor predating the protostomes split from deuterostomes approximately 550 million years ago.


Subject(s)
Calcium Channels, L-Type/genetics , Drosophila Proteins/genetics , Evolution, Molecular , Monomeric GTP-Binding Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , ras Proteins/genetics , Amino Acid Sequence , Animals , Calcium Channels, L-Type/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Molecular Sequence Data , Monomeric GTP-Binding Proteins/metabolism , Rats , Rats, Wistar , Zebrafish/metabolism , Zebrafish Proteins/metabolism , ras Proteins/metabolism
11.
J Neurosci ; 33(49): 19314-25, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305827

ABSTRACT

Free fatty acids receptor 3 (FFA3, GPR41) and 2 (FFA2, GPR43), for which the short-chain fatty acids (SCFAs) acetate and propionate are agonist, have emerged as important G-protein-coupled receptors influenced by diet and gut flora composition. A recent study (Kimura et al., 2011) demonstrated functional expression of FFA3 in the rodent sympathetic nervous system (SNS) providing a potential link between nutritional status and autonomic function. However, little is known of the source of endogenous ligands, signaling pathways, or effectors in sympathetic neurons. In this study, we found that FFA3 and FFA2 are unevenly expressed in the rat SNS with higher transcript levels in prevertebral (e.g., celiac-superior mesenteric and major pelvic) versus paravertebral (e.g., superior cervical and stellate) ganglia. FFA3, whether heterologously or natively expressed, coupled via PTX-sensitive G-proteins to produce voltage-dependent inhibition of N-type Ca(2+) channels (Cav2.2) in sympathetic neurons. In addition to acetate and propionate, we show that ß-hydroxybutyrate (BHB), a metabolite produced during ketogenic conditions, is also an FFA3 agonist. This contrasts with previous interpretations of BHB as an antagonist at FFA3. Together, these results indicate that endogenous BHB levels, especially when elevated under certain conditions, such as starvation, diabetic ketoacidosis, and ketogenic diets, play a potentially important role in regulating the activity of the SNS through FFA3.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Calcium Channels, N-Type/drug effects , Neurons/physiology , Receptors, G-Protein-Coupled/agonists , Sympathetic Nervous System/physiology , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiological Phenomena/physiology , Fluorescence Resonance Energy Transfer , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/physiology , HeLa Cells , Humans , In Situ Hybridization , Ketone Bodies/pharmacology , Ligands , Male , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sympathetic Nervous System/cytology , Transfection
12.
Mol Pharmacol ; 83(1): 267-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23104136

ABSTRACT

Recent studies propose that N-arachidonyl glycine (NAGly), a carboxylic analogue of anandamide, is an endogenous ligand of the Gα(i/o) protein-coupled receptor 18 (GPR18). However, a high-throughput ß-arrestin-based screen failed to detect activation of GPR18 by NAGly (Yin et al., 2009; JBC, 18:12328). To address this inconsistency, this study investigated GPR18 coupling in a native neuronal system with endogenous signaling pathways and effectors. GPR18 was heterologously expressed in rat sympathetic neurons, and the modulation of N-type (Ca(v)2.2) calcium channels was examined. Proper expression and trafficking of receptor were confirmed by the "rim-like" fluorescence of fluorescently tagged receptor and the positive staining of external hemagglutinin-tagged GPR18-expressing cells. Application of NAGly on GPR18-expressing neurons did not inhibit calcium currents but instead potentiated currents in a voltage-dependent manner, similar to what has previously been reported (Guo et al., 2008; J Neurophysiol, 100:1147). Other proposed agonists of GPR18, including anandamide and abnormal cannabidiol, also failed to induce inhibition of calcium currents. Mutants of GPR18, designed to constitutively activate receptors, did not tonically inhibit calcium currents, indicating a lack of GPR18 activation or coupling to endogenous G proteins. Other downstream effectors of Gα(i/o)-coupled receptors, G protein-coupled inwardly rectifying potassium channels and adenylate cyclase, were not modulated by GPR18 signaling. Furthermore, GPR18 did not couple to other G proteins tested: Gα(s), Gα(z), and Gα(15). These results suggest NAGly is not an agonist for GPR18 or that GPR18 signaling involves noncanonical pathways not examined in these studies.


Subject(s)
Arachidonic Acids/pharmacology , Glycine/analogs & derivatives , Receptors, G-Protein-Coupled/agonists , Animals , Calcium Channels, N-Type/physiology , Cell Membrane/metabolism , Cyclic AMP/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Glycine/pharmacology , HEK293 Cells , HeLa Cells , Humans , In Vitro Techniques , Male , Mutation , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Superior Cervical Ganglion/cytology
13.
PLoS One ; 7(8): e42602, 2012.
Article in English | MEDLINE | ID: mdl-22880050

ABSTRACT

BACKGROUND: Dorsal root ganglia (DRG) somata from rodents have provided an excellent model system to study ion channel properties and modulation using electrophysiological investigation. As in other vertebrates, zebrafish (Danio rerio) DRG are organized segmentally and possess peripheral axons that bifurcate into each body segment. However, the electrical properties of zebrafish DRG sensory neurons, as compared with their mammalian counterparts, are relatively unexplored because a preparation suitable for electrophysiological studies has not been available. METHODOLOGY/PRINCIPAL FINDINGS: We show enzymatically dissociated DRG neurons from juvenile zebrafish expressing Isl2b-promoter driven EGFP were easily identified with fluorescence microscopy and amenable to conventional whole-cell patch-clamp studies. Two kinetically distinct TTX-sensitive Na(+) currents (rapidly- and slowly-inactivating) were discovered. Rapidly-inactivating I(Na) were preferentially expressed in relatively large neurons, while slowly-inactivating I(Na) was more prevalent in smaller DRG neurons. RT-PCR analysis suggests zscn1aa/ab, zscn8aa/ab, zscn4ab and zscn5Laa are possible candidates for these I(Na) components. Voltage-gated Ca(2+) currents (I(Ca)) were primarily (87%) comprised of a high-voltage activated component arising from ω-conotoxin GVIA-sensitive Ca(V)2.2 (N-type) Ca(2+) channels. A few DRG neurons (8%) displayed a miniscule low-voltage-activated component. I(Ca) in zebrafish DRG neurons were modulated by neurotransmitters via either voltage-dependent or -independent G-protein signaling pathway with large cell-to-cell response variability. CONCLUSIONS/SIGNIFICANCE: Our present results indicate that, as in higher vertebrates, zebrafish DRG neurons are heterogeneous being composed of functionally distinct subpopulations that may correlate with different sensory modalities. These findings provide the first comparison of zebrafish and rodent DRG neuron electrical properties and thus provide a basis for future studies.


Subject(s)
Calcium Channels/metabolism , Ganglia, Spinal/cytology , Neurons/metabolism , Sodium Channels/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Calcium Channels/genetics , GTP-Binding Proteins/metabolism , Ganglia, Spinal/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Ion Channel Gating , Kinetics , Molecular Sequence Data , Neurons/cytology , Neurotransmitter Agents/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA-B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
J Neurophysiol ; 105(1): 442-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962070

ABSTRACT

Electrically excitable cells have voltage-dependent ion channels on the plasma membrane that regulate membrane permeability to specific ions. Voltage-gated Ca(2+) channels (VGCCs) are especially important as Ca(2+) serves as both a charge carrier and second messenger. Zebrafish (Danio rerio) are an important model vertebrate for studies of neuronal excitability, circuits, and behavior. However, electrophysiological properties of zebrafish VGCCs remain largely unexplored because a suitable preparation for whole cell voltage-clamp studies is lacking. Rohon-Beard (R-B) sensory neurons represent an attractive candidate for this purpose because of their relatively large somata and functional homology to mammalian dorsal root ganglia (DRG) neurons. Transgenic zebrafish expressing green fluorescent protein in R-B neurons, (Isl2b:EGFP)(ZC7), were used to identify dissociated neurons suitable for whole cell patch-clamp experiments. Based on biophysical and pharmacological properties, zebrafish R-B neurons express both high- and low-voltage-gated Ca(2+) current (HVA- and LVA-I(Ca), respectively). Ni(+)-sensitive LVA-I(Ca) occur in the minority of R-B neurons (30%) and ω-conotoxin GVIA-sensitive Ca(V)2.2 (N-type) Ca(2+) channels underlie the vast majority (90%) of HVA-I(Ca). To identify G protein coupled receptors (GPCRs) that modulate HVA-I(Ca), a panel of neurotransmitters was screened. Application of GABA/baclofen or serotonin produced a voltage-dependent inhibition while application of the mu-opioid agonist DAMGO resulted in a voltage-independent inhibition. Unlike in mammalian neurons, GPCR-mediated voltage-dependent modulation of I(Ca) appears to be transduced primarily via a cholera toxin-sensitive Gα subunit. These results provide the basis for using the zebrafish model system to understanding Ca(2+) channel function, and in turn, how Ca(2+) channels contribute to mechanosensory function.


Subject(s)
Calcium Channels/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Baclofen/pharmacology , Calcium Channels/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Green Fluorescent Proteins/genetics , Models, Animal , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , gamma-Aminobutyric Acid/pharmacology
15.
Front Neurosci ; 4: 181, 2010.
Article in English | MEDLINE | ID: mdl-21267423

ABSTRACT

Expression of heterologous proteins in adult mammalian neurons is a valuable technique for the study of neuronal function. The post-mitotic nature of mature neurons prevents effective DNA transfection using simple, cationic lipid-based methods. Adequate heterologous protein expression is often only achievable using complex techniques that, in many cases, are associated with substantial toxicity. Here, a simple method for high efficiency transfection of mammalian primary neurons using in vitro transcribed mRNA and the cationic lipid transfection reagent Lipofectamine™ 2000 is described. Optimal transfection conditions were established in adult mouse dissociated dorsal root ganglion (DRG) neurons using a 96-well based luciferase activity assay. Using these conditions, a transfection efficiency of 25% was achieved in DRG neurons transfected with EGFP mRNA. High transfection efficiencies were also obtained in dissociated rat superior cervical ganglion (SCG) neurons and mouse cortical and hippocampal cultures. Endogenous Ca(2+) currents in EGFP mRNA-transfected SCG neurons were not significantly different from untransfected neurons, which suggested that this technique is well suited for heterologous expression in patch clamp recording experiments. Functional expression of a cannabinoid receptor (CB1R), a G protein inwardly rectifying K(+) channel (GIRK4) and a dominant-negative G protein α-subunit mutant (G(oA) G203T) indicate that the levels of heterologous protein expression attainable using mRNA transfection are suitable for most functional protein studies. This study demonstrates that mRNA transfection is a straightforward and effective method for heterologous expression in neurons and is likely to have many applications in neuroscience research.

16.
J Vis Exp ; (34)2009 Dec 10.
Article in English | MEDLINE | ID: mdl-20010540

ABSTRACT

Primary neuronal cell cultures are valuable tools to study protein function since they represent a more biologically relevant system compared to immortalized cell lines. However, the post-mitotic nature of primary neurons prevents effective heterologous protein expression using common procedures such as electroporation or chemically-mediated transfection. Thus, other techniques must be employed in order to effectively express proteins in these non-dividing cells. In this article, we describe the steps required to perform intranuclear injections of cDNA constructs into dissociated adult sympathetic neurons. This technique, which has been applied to different types of neurons, can successfully induce heterologous protein expression. The equipment essential for the microinjection procedure includes an inverted microscope to visualize cells, a glass injection pipet filled with cDNA solution that is connected to a N2(g) pressure delivery system, and a micromanipulator. The micromanipulator coordinates the injection motion of microinjection pipet with a brief pulse of pressurized N2 to eject cDNA solution from the pipet tip. This technique does not have the toxicity associated with many other transfection methods and enables multiple DNA constructs to be expressed at a consistent ratio. The low number of injected cells makes the microinjection procedure well suited for single cell studies such as electrophysiological recordings and optical imaging, but may not be ideal for biochemical assays that require a larger number of cells and higher transfection efficiencies. Although intranuclear microinjections require an investment of equipment and time, the ability to achieve high levels of heterologous protein expression in a physiologically relevant environment makes this technique a very useful tool to investigate protein function.


Subject(s)
DNA, Complementary/administration & dosage , Microinjections/methods , Neurons/physiology , Sympathetic Nervous System/cytology , Transfection/methods , Adult , Humans , Intranuclear Space
17.
J Neurosci ; 29(43): 13603-12, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19864572

ABSTRACT

Endocannabinoids (eCB) such as 2-arachidonylglycerol (2-AG) are lipid metabolites that are synthesized in a postsynaptic neurons and act upon CB(1) cannabinoid receptors (CB(1)R) in presynaptic nerve terminals. This retrograde transmission underlies several forms of short and long term synaptic plasticity within the CNS. Here, we constructed a model system based on isolated rat sympathetic neurons, in which an eCB signaling cascade could be studied in a reduced, spatially compact, and genetically malleable system. We constructed a complete eCB production/mobilization pathway by sequential addition of molecular components. Heterologous expression of four components was required for eCB production and detection: metabotropic glutamate receptor 5a (mGluR5a), Homer 2b, diacylglycerol lipase alpha, and CB(1)R. In these neurons, application of l-glutamate produced voltage-dependent modulation of N-type Ca(2+) channels mediated by activation of CB(1)R. Using both molecular dissection and pharmacological agents, we provide evidence that activation of mGluR5a results in rapid enzymatic production of 2-AG followed by activation of CB(1)R. These experiments define the critical elements required to recapitulate retrograde eCB production and signaling in a single peripheral neuron. Moreover, production/mobilization of eCB can be detected on a physiologically relevant time scale using electrophysiological techniques. The system provides a platform for testing candidate molecules underlying facilitation of eCB transport across the plasma membrane.


Subject(s)
Arachidonic Acids/metabolism , Glycerides/metabolism , Models, Neurological , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Superior Cervical Ganglion/metabolism , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Endocannabinoids , Glutamic Acid/metabolism , Homer Scaffolding Proteins , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Male , Patch-Clamp Techniques , Peripheral Nerves/metabolism , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Metabotropic Glutamate 5 , Signal Transduction , Time Factors
18.
PLoS One ; 4(10): e7474, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19830250

ABSTRACT

BACKGROUND: The ability to disrupt the function of a specific protein on a rapid time scale provides a powerful tool for biomedical research. Specific proteases provide a potential method to selectively cleave a chosen protein, but rapid control of protease activity is difficult. METHODOLOGY/PRINCIPAL FINDINGS: A heterologous expression system for rapid target-directed proteolysis in mammalian cells was developed. The system consists of an inducible NIa protease from the tobacco etch virus (TEVp) and a chosen protein into which a TEVp substrate recognition sequence (TRS) has been inserted. Inducible activity was conferred to the TEVp using rapamycin-controlled TEVp fragment complementation. TEVp activity was assayed using a FRET-based reporter construct. TEVp expression was well tolerated by mammalian cells and complete cleavage of the substrate was possible. Cleavage at 37 degrees C proceeded exponentially with a time constant of approximately 150 minutes. Attempts to improve cleavage efficiency were hampered by substantial background activity, which was attributed to inherent affinity between the TEVp fragments. A second TEVp assay, based on changes in inactivation of a modified K(V)3.4 channel, showed that functional properties of a channel can be using altered using an inducible TEVp system. Similar levels of background activity and variability were observed in both electrophysiological and FRET assays. CONCLUSIONS/SIGNIFICANCE: The results suggested that an optimum level of TEVp expression leading to sufficient inducible activity (with minimal background activity) exists but the variability in expression levels between cells makes the present system rather impractical for single cell experiments. The system is likely to be more suitable for experiments in which the cell-to-cell variability is less of an issue; for example, in experiments involving large populations of cells.


Subject(s)
Endopeptidases/chemistry , Genetic Techniques , Potyvirus/metabolism , Protein Engineering/methods , Proteins/chemistry , Sirolimus/chemistry , Cell Line , Electrophysiology/methods , Fluorescence Resonance Energy Transfer/methods , Humans , Kinetics , Models, Chemical , Shaw Potassium Channels/chemistry , Temperature , Time Factors
19.
J Biol Chem ; 283(52): 36698-710, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18984596

ABSTRACT

Heterotrimeric G-protein Galpha subunits and GoLoco motif proteins are key members of a conserved set of regulatory proteins that influence invertebrate asymmetric cell division and vertebrate neuroepithelium and epithelial progenitor differentiation. GoLoco motif proteins bind selectively to the inhibitory subclass (Galphai) of Galpha subunits, and thus it is assumed that a Galphai.GoLoco motif protein complex plays a direct functional role in microtubule dynamics underlying spindle orientation and metaphase chromosomal segregation during cell division. To address this hypothesis directly, we rationally identified a point mutation to Galphai subunits that renders a selective loss-of-function for GoLoco motif binding, namely an asparagine-to-isoleucine substitution in the alphaD-alphaE loop of the Galpha helical domain. This GoLoco-insensitivity ("GLi") mutation prevented Galphai1 association with all human GoLoco motif proteins and abrogated interaction between the Caenorhabditis elegans Galpha subunit GOA-1 and the GPR-1 GoLoco motif. In contrast, the GLi mutation did not perturb any other biochemical or signaling properties of Galphai subunits, including nucleotide binding, intrinsic and RGS protein-accelerated GTP hydrolysis, and interactions with Gbetagamma dimers, adenylyl cyclase, and seven transmembrane-domain receptors. GoLoco insensitivity rendered Galphai subunits unable to recruit GoLoco motif proteins such as GPSM2/LGN and GPSM3 to the plasma membrane, and abrogated the exaggerated mitotic spindle rocking normally seen upon ectopic expression of wild type Galphai subunits in kidney epithelial cells. This GLi mutation should prove valuable in establishing the physiological roles of Galphai.GoLoco motif protein complexes in microtubule dynamics and spindle function during cell division as well as to delineate potential roles for GoLoco motifs in receptor-mediated signal transduction.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Spindle Apparatus , Amino Acid Motifs , Amino Acid Sequence , Animals , Caenorhabditis elegans , Cell Membrane/metabolism , Humans , Microtubules/metabolism , Models, Molecular , Molecular Sequence Data , Point Mutation , Rats , Sequence Homology, Amino Acid , Signal Transduction
20.
J Neurochem ; 106(3): 1209-24, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18466327

ABSTRACT

Voltage-gated sodium channels (VGSC) are critical membrane components that participate in the electrical activity of excitable cells. The type one VGSC family includes the tetrodotoxin insensitive sodium channel, Na(V)1.8, encoded by the Scn10a gene. Na(V)1.8 expression is restricted to small and medium diameter nociceptive sensory neurons of the dorsal root ganglia and cranial sensory ganglia. To understand the stringent transcriptional regulation of the Scn10a gene, the sensory neuron specific promoter was functionally identified. While identifying the mRNA 5'-end, alternative splicing within the 5'-UTR was observed to create heterogeneity in the RNA transcript. Four kilobases of upstream genomic DNA was cloned and the presence of tissue specific promoter activity was tested by microinjection and adenoviral infection of fluorescent protein reporter constructs into primary mouse and rat neurons, and cell lines. The region contained many putative transcription factor-binding sites and strong homology with the predicted rat ortholog. Homology to the predicted human ortholog was limited to the proximal end and several conserved cis elements were noted. Two regulatory modules were identified by microinjection of reporter constructs into dorsal root ganglia and superior cervical ganglia neurons: a neuron specific proximal promoter region between -1.6 and -0.2 kb of the transcription start site cluster, and a distal sensory neuron switch region beyond -1.6 kb that restricted fluorescent protein expression to a subset of primary sensory neurons.


Subject(s)
Neurons, Afferent/physiology , Regulatory Sequences, Nucleic Acid/genetics , Sodium Channels/genetics , 5' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Ganglia, Spinal/physiology , HeLa Cells , Humans , Male , Mice , Molecular Sequence Data , NAV1.8 Voltage-Gated Sodium Channel , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sodium Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...