Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Neuroendocrinol ; 35(11): e13255, 2023 11.
Article in English | MEDLINE | ID: mdl-36970756

ABSTRACT

Adrenomedullary chromaffin cells respond to splanchnic (sympathetic) nerve stimulation by releasing stress hormones into the circulation. The signal for hormone secretion is encoded in the neurotransmitters - especially acetylcholine (ACh) and pituitary adenylate cyclase activating polypeptide (PACAP) - that are released into the splanchnic-chromaffin cell synapse. However, functional differences in the effects of ACh and PACAP on the chromaffin cell secretory response are not well defined. Here, selective agonists of PACAP receptors or nicotinic and muscarinic acetylcholine receptors were applied to chromaffin cells. The major differences in the effects of these agents were not on exocytosis, per se, but rather on the steps upstream of exocytosis. In almost every respect, the properties of individual fusion events triggered by PACAP and cholinergic agonists were similar. On the other hand, the properties of the Ca2+ transients evoked by PACAP differed in several ways from those evoked by muscarinic and nicotinic receptor stimulation. A defining feature of the PACAP-stimulated secretory pathway was its dependence on signaling through exchange protein directly activated by cAMP (Epac) and PLCε. However, the absence of PLCε did not disrupt Ca2+ transients evoked by cholinergic agonists. Accordingly, inhibition of Epac activity did not disrupt secretion triggered by acetylcholine or specific agonists of muscarinic and nicotinic receptors. Thus, PACAP and acetylcholine stimulate chromaffin cell secretion via separate and independent pathways. This feature of stimulus-secretion coupling may be important for sustaining hormone release from the adrenal medulla under conditions associated with the sympathetic stress response.


Subject(s)
Chromaffin Cells , Pituitary Adenylate Cyclase-Activating Polypeptide , Acetylcholine/metabolism , Catecholamines/metabolism , Catecholamines/pharmacology , Cholinergic Agonists/metabolism , Cholinergic Agonists/pharmacology , Chromaffin Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hormones , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Mice , Receptors, Cholinergic/metabolism
2.
Sci Rep ; 10(1): 1300, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992767

ABSTRACT

The dorsal root ganglia (DRG) house the primary afferent neurons responsible for somatosensation, including pain. We previously identified Jedi-1 (PEAR1/MEGF12) as a phagocytic receptor expressed by satellite glia in the DRG involved in clearing apoptotic neurons during development. Here, we further investigated the function of this receptor in vivo using Jedi-1 null mice. In addition to satellite glia, we found Jedi-1 expression in perineurial glia and endothelial cells, but not in sensory neurons. We did not detect any morphological or functional changes in the glial cells or vasculature of Jedi-1 knockout mice. Surprisingly, we did observe changes in DRG neuron activity. In neurons from Jedi-1 knockout (KO) mice, there was an increase in the fraction of capsaicin-sensitive cells relative to wild type (WT) controls. Patch-clamp electrophysiology revealed an increase in excitability, with a shift from phasic to tonic action potential firing patterns in KO neurons. We also found alterations in the properties of voltage-gated sodium channel currents in Jedi-1 null neurons. These results provide new insight into the expression pattern of Jedi-1 in the peripheral nervous system and indicate that loss of Jedi-1 alters DRG neuron activity indirectly through an intercellular interaction between non-neuronal cells and sensory neurons.


Subject(s)
Action Potentials , Receptors, Cell Surface/deficiency , Sensory Receptor Cells/metabolism , Animals , Biomarkers , Cell Line , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Neuroglia/metabolism , Neuroglia/ultrastructure , Patch-Clamp Techniques , Sensory Receptor Cells/ultrastructure
3.
Pharmacol Res ; 140: 56-66, 2019 02.
Article in English | MEDLINE | ID: mdl-29894763

ABSTRACT

Adrenal chromaffin cells comprise the neuroendocrine arm of the sympathetic nervous system and secrete catecholamines to coordinate the appropriate stress response. Deletion of the serotonin (5-HT) transporter (SERT) gene in mice (SERT-/- mice) or pharmacological block of SERT function in rodents and humans augments this sympathoadrenal stress response (epinephrine secretion). The prevailing assumption is that loss of CNS SERT alters central drive to the peripheral sympathetic nervous system. Adrenal chromaffin cells also prominently express SERT where it might coordinate accumulation of 5-HT for reuse in the autocrine control of stress-evoked catecholamine secretion. To help test this hypothesis, we have generated a novel mouse model with selective excision of SERT in the peripheral sympathetic nervous system (SERTΔTH), generated by crossing floxed SERT mice with tyrosine hydroxylase Cre driver mice. SERT expression, assessed by western blot, was abolished in the adrenal gland but not perturbed in the CNS of SERTΔTH mice. SERT-mediated [3H] 5-HT uptake was unaltered in midbrain, hindbrain, and spinal cord synaptosomes, confirming transporter function was intact in the CNS. Endogenous midbrain and whole blood 5-HT homeostasis was unperturbed in SERTΔTH mice, contrasting with the depleted 5-HT content in SERT-/- mice. Selective SERT excision reduced adrenal gland 5-HT content by ≈ 50% in SERTΔTH mice but had no effect on adrenal catecholamine content. This novel model confirms that SERT expressed in adrenal chromaffin cells is essential for maintaining wild-type levels of 5-HT and provides a powerful tool to help dissect the role of SERT in the sympathetic stress response.


Subject(s)
Adrenal Glands/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Antidepressive Agents , Female , Male , Mesencephalon/metabolism , Mice, Transgenic , Models, Animal , Rhombencephalon/metabolism , Serotonin/metabolism , Spinal Cord/metabolism , Tyrosine 3-Monooxygenase
4.
J Neurochem ; 143(2): 171-182, 2017 10.
Article in English | MEDLINE | ID: mdl-28815595

ABSTRACT

Adrenal chromaffin cells (ACCs) are the neuroendocrine arm of the sympathetic nervous system and key mediators of the physiological stress response. Acetylcholine (ACh) released from preganglionic splanchnic nerves activates nicotinic acetylcholine receptors (nAChRs) on chromaffin cells causing membrane depolarization, opening voltage-gated Ca2+ channels (VGCC), and exocytosis of catecholamines and neuropeptides. The serotonin transporter is expressed in ACCs and interacts with 5-HT1A receptors to control secretion. In addition to blocking the serotonin transporter, some selective serotonin reuptake inhibitors (SSRIs) are also agonists at sigma-1 receptors which function as intracellular chaperone proteins and can translocate to the plasma membrane to modulate ion channels. Therefore, we investigated whether SSRIs and other sigma-1 receptor ligands can modulate stimulus-secretion coupling in ACCs. Escitalopram and fluvoxamine (100 nM to 1 µM) reversibly inhibited nAChR currents. The sigma-1 receptor antagonists NE-100 and BD-1047 also blocked nAChR currents (≈ 50% block at 100 nM) as did PRE-084, a sigma-1 receptor agonist. Block of nAChR currents by fluvoxamine and NE-100 was not additive suggesting a common site of action. VGCC currents were unaffected by the drugs. Neither the increase in cytosolic [Ca2+ ] nor the resulting catecholamine secretion evoked by direct membrane depolarization to bypass nAChRs was altered by fluvoxamine or NE-100. However, both Ca2+ entry and catecholamine secretion evoked by the cholinergic agonist carbachol were significantly reduced by fluvoxamine or NE-100. Together, our data suggest that sigma-1 receptors do not acutely regulate catecholamine secretion. Rather, SSRIs and other sigma-1 receptor ligands inhibit secretion evoked by cholinergic stimulation because of direct block of Ca2+ entry via nAChRs.


Subject(s)
Adrenal Medulla/metabolism , Catecholamines/metabolism , Chromaffin Cells/metabolism , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/physiology , Receptors, sigma/physiology , Adrenal Medulla/cytology , Adrenal Medulla/drug effects , Animals , Anisoles/pharmacology , Catecholamines/antagonists & inhibitors , Cattle , Cells, Cultured , Chromaffin Cells/drug effects , Dose-Response Relationship, Drug , Ligands , Male , Mice , Mice, Inbred C57BL , Propylamines/pharmacology , Receptors, sigma/agonists , Sigma-1 Receptor
5.
J Biol Chem ; 292(29): 12165-12177, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28515322

ABSTRACT

Gi/o-coupled G protein-coupled receptors can inhibit neurotransmitter release at synapses via multiple mechanisms. In addition to Gßγ-mediated modulation of voltage-gated calcium channels (VGCC), inhibition can also be mediated through the direct interaction of Gßγ subunits with the soluble N-ethylmaleimide attachment protein receptor (SNARE) complex of the vesicle fusion apparatus. Binding studies with soluble SNARE complexes have shown that Gßγ binds to both ternary SNARE complexes, t-SNARE heterodimers, and monomeric SNAREs, competing with synaptotagmin 1(syt1) for binding sites on t-SNARE. However, in secretory cells, Gßγ, SNAREs, and synaptotagmin interact in the lipid environment of a vesicle at the plasma membrane. To approximate this environment, we show that fluorescently labeled Gßγ interacts specifically with lipid-embedded t-SNAREs consisting of full-length syntaxin 1 and SNAP-25B at the membrane, as measured by fluorescence polarization. Fluorescently labeled syt1 undergoes competition with Gßγ for SNARE-binding sites in lipid environments. Mutant Gßγ subunits that were previously shown to be more efficacious at inhibiting Ca2+-triggered exocytotic release than wild-type Gßγ were also shown to bind SNAREs at a higher affinity than wild type in a lipid environment. These mutant Gßγ subunits were unable to inhibit VGCC currents. Specific peptides corresponding to regions on Gß and Gγ shown to be important for the interaction disrupt the interaction in a concentration-dependent manner. In in vitro fusion assays using full-length t- and v-SNAREs embedded in liposomes, Gßγ inhibited Ca2+/synaptotagmin-dependent fusion. Together, these studies demonstrate the importance of these regions for the Gßγ-SNARE interaction and show that the target of Gßγ, downstream of VGCC, is the membrane-embedded SNARE complex.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Lipid Bilayers , Models, Molecular , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Syntaxin 1/metabolism , Animals , Binding, Competitive , Calcium Signaling , Cattle , Cell Line , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , Humans , Liposomes , Membrane Fusion , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Synaptosomal-Associated Protein 25/chemistry , Synaptotagmin I/chemistry , Synaptotagmin I/genetics , Syntaxin 1/chemistry
6.
ACS Chem Neurosci ; 8(5): 943-954, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28406285

ABSTRACT

Serotonin (5-HT) is an important neurotransmitter in the central nervous system where it modulates circuits involved in mood, cognition, movement, arousal, and autonomic function. The 5-HT transporter (SERT; SLC6A4) is a key regulator of 5-HT signaling, and genetic variations in SERT are associated with various disorders including depression, anxiety, and autism. This review focuses on the role of SERT in the sympathetic nervous system. Autonomic/sympathetic dysfunction is evident in patients with depression, anxiety, and other diseases linked to serotonergic signaling. Experimentally, loss of SERT function (SERT knockout mice or chronic pharmacological block) has been reported to augment the sympathetic stress response. Alterations to serotonergic signaling in the CNS and thus central drive to the peripheral sympathetic nervous system are presumed to underlie this augmentation. Although less widely recognized, SERT is robustly expressed in chromaffin cells of the adrenal medulla, the neuroendocrine arm of the sympathetic nervous system. Adrenal chromaffin cells do not synthesize 5-HT but accumulate small amounts by SERT-mediated uptake. Recent evidence demonstrated that 5-HT1A receptors inhibit catecholamine secretion from adrenal chromaffin cells via an atypical mechanism that does not involve modulation of cellular excitability or voltage-gated Ca2+ channels. This raises the possibility that the adrenal medulla is a previously unrecognized peripheral hub for serotonergic control of the sympathetic stress response. As a framework for future investigation, a model is proposed in which stress-evoked adrenal catecholamine secretion is fine-tuned by SERT-modulated autocrine 5-HT signaling.


Subject(s)
Adrenal Medulla/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Stress, Physiological/physiology , Sympathetic Nervous System/metabolism , Animals , Arousal/physiology , Chromaffin Cells/metabolism , Humans , Synaptic Transmission/physiology
7.
Neuropharmacology ; 110(Pt A): 438-448, 2016 11.
Article in English | MEDLINE | ID: mdl-27544824

ABSTRACT

Adrenal chromaffin cells (ACCs), the neuroendocrine arm of the sympathetic nervous system, secrete catecholamines to mediate the physiological response to stress. Although ACCs do not synthesize 5-HT, they express the serotonin transporter (SERT). Genetic variations in SERT are linked to several CNS disorders but the role(s) of SERT/5-HT in ACCs has remained unclear. Adrenal glands from wild-type mice contained 5-HT at ≈ 750 fold lower abundance than adrenaline, and in SERT(-/-) mice this was reduced by ≈80% with no change in catecholamines. Carbon fibre amperometry showed that SERT modulated the ability of 5-HT1A receptors to inhibit exocytosis. 5-HT reduced the number of amperometric spikes (vesicular fusion events) evoked by KCl in SERT(-/-) cells and wild-type cells treated with escitalopram, a SERT antagonist. The 5-HT1A receptor antagonist WAY100635 blocked the inhibition by 5-HT which was mimicked by the 5-HT1A agonist 8-OH-DPAT but not the 5-HT1B agonist CP93129. There was no effect on voltage-gated Ca(2+) channels, K(+) channels, or intracellular [Ca(2+)] handling, showing the 5-HT receptors recruit an atypical inhibitory mechanism. Spike charge and kinetics were not altered by 5-HT receptors but were reduced in SERT(-/-) cells compared to wild-type cells. Our data reveal a novel role for SERT and suggest that adrenal chromaffin cells might be a previously unrecognized hub for serotonergic control of the sympathetic stress response.


Subject(s)
Adrenal Glands/metabolism , Chromaffin Cells/metabolism , Receptors, Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Adrenal Glands/drug effects , Animals , Calcium/metabolism , Calcium Channels, N-Type/metabolism , Cations, Divalent/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Exocytosis/drug effects , Exocytosis/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Voltage-Gated/metabolism , Serotonin/metabolism , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics
8.
PLoS One ; 9(10): e109203, 2014.
Article in English | MEDLINE | ID: mdl-25275439

ABSTRACT

Butanol (C4H10OH) has been used both to dissect the molecular targets of alcohols/general anesthetics and to implicate phospholipase D (PLD) signaling in a variety of cellular functions including neurotransmitter and hormone exocytosis. Like other primary alcohols, 1-butanol is a substrate for PLD and thereby disrupts formation of the intracellular signaling lipid phosphatidic acid. Because secondary and tertiary butanols do not undergo this transphosphatidylation, they have been used as controls for 1-butanol to implicate PLD signaling. Recently, selective pharmacological inhibitors of PLD have been developed and, in some cases, fail to block cellular functions previously ascribed to PLD using primary alcohols. For example, exocytosis of insulin and degranulation of mast cells are blocked by primary alcohols, but not by the PLD inhibitor FIPI. In this study we show that 1-butanol reduces catecholamine secretion from adrenal chromaffin cells to a much greater extent than tert-butanol, and that the PLD inhibitor VU0155056 has no effect. Using fluorescent imaging we show the effect of these drugs on depolarization-evoked calcium entry parallel those on secretion. Patch-clamp electrophysiology confirmed the peak amplitude of voltage-gated calcium channel currents (I(Ca)) is inhibited by 1-butanol, with little or no block by secondary or tert-butanol. Detailed comparison shows for the first time that the different butanol isomers exert distinct, and sometimes opposing, effects on the voltage-dependence and gating kinetics of I(Ca). We discuss these data with regard to PLD signaling in cellular physiology and the molecular targets of general anesthetics.


Subject(s)
Butanols/pharmacology , Calcium Channels/drug effects , Catecholamines/metabolism , Chromaffin Cells/drug effects , Ion Channel Gating , Animals , Butanols/chemistry , Calcium/metabolism , Cattle , Chromaffin Cells/metabolism , Isomerism
9.
Lab Chip ; 13(23): 4663-73, 2013 Dec 07.
Article in English | MEDLINE | ID: mdl-24126415

ABSTRACT

Release of neurotransmitters and hormones by calcium-regulated exocytosis is a fundamental cellular process that is disrupted in a variety of psychiatric, neurological, and endocrine disorders. As such, there is significant interest in targeting neurosecretion for drug and therapeutic development, efforts that will be aided by novel analytical tools and devices that provide mechanistic insight coupled with increased experimental throughput. Here, we report a simple, inexpensive, reusable, microfluidic device designed to analyze catecholamine secretion from small populations of adrenal chromaffin cells in real time, an important neuroendocrine component of the sympathetic nervous system and versatile neurosecretory model. The device is fabricated by replica molding of polydimethylsiloxane (PDMS) using patterned photoresist on silicon wafer as the master. Microfluidic inlet channels lead to an array of U-shaped "cell traps", each capable of immobilizing single or small groups of chromaffin cells. The bottom of the device is a glass slide with patterned thin film platinum electrodes used for electrochemical detection of catecholamines in real time. We demonstrate reliable loading of the device with small populations of chromaffin cells, and perfusion/repetitive stimulation with physiologically relevant secretagogues (carbachol, PACAP, KCl) using the microfluidic network. Evoked catecholamine secretion was reproducible over multiple rounds of stimulation, and graded as expected to different concentrations of secretagogue or removal of extracellular calcium. Overall, we show this microfluidic device can be used to implement complex stimulation paradigms and analyze the amount and kinetics of catecholamine secretion from small populations of neuroendocrine cells in real time.


Subject(s)
Catecholamines/analysis , Chromaffin Cells/metabolism , Chromatography, High Pressure Liquid , Electrochemical Techniques , Microfluidic Analytical Techniques/instrumentation , Animals , Calcium/metabolism , Carbachol/pharmacology , Catecholamines/metabolism , Cattle , Cells, Cultured , Chromaffin Cells/cytology , Chromaffin Cells/drug effects , Dimethylpolysiloxanes , Electrodes , Kinetics , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Platinum/chemistry , Potassium Chloride/pharmacology , Silicon/chemistry , Stimulation, Chemical
10.
Anesthesiology ; 116(5): 1013-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22417967

ABSTRACT

BACKGROUND: Gabapentin is most commonly prescribed for chronic pain, but acute perioperative effects, including preemptive analgesia and hemodynamic stabilization, have been reported. Adrenal chromaffin cells are a widely used model to investigate neurosecretion, and adrenal catecholamines play important physiologic roles and contribute to the acute stress response. However, the effects of gabapentin on adrenal catecholamine release have never been tested. METHODS: Primary cultures of bovine adrenal chromaffin cells were treated with gabapentin or vehicle for 18-24 h. The authors quantified catecholamine secretion from dishes of cells using high-performance liquid chromatography and resolved exocytosis of individual secretory vesicles from single cells using carbon fiber amperometry. Voltage-gated calcium channel currents were recorded using patch clamp electrophysiology and intracellular [Ca2+] using fluorescent imaging. RESULTS: Gabapentin produced statistically significant reductions in catecholamine secretion evoked by cholinergic agonists (24 ± 3%, n = 12) or KCl (16 ± 4%, n = 8) (mean ± SEM) but did not inhibit Ca2+ entry or calcium channel currents. Amperometry (n = 51 cells) revealed that gabapentin inhibited the number of vesicles released upon stimulation, with no change in quantal size or kinetics of these unitary events. CONCLUSIONS: The authors show Ca2+ entry was not inhibited by gabapentin but was less effective at triggering vesicle fusion. The work also demonstrates that chromaffin cells are a useful model for additional investigation of the cellular mechanism(s) by which gabapentin controls neurosecretion. In addition, it identifies altered adrenal catecholamine release as a potential contributor to some of the beneficial perioperative effects of gabapentin.


Subject(s)
Adrenal Glands/metabolism , Amines/pharmacology , Catecholamines/antagonists & inhibitors , Catecholamines/metabolism , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , gamma-Aminobutyric Acid/pharmacology , Adrenal Glands/cytology , Adrenal Glands/drug effects , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Signaling/drug effects , Cattle , Cholinergic Agonists/pharmacology , Dose-Response Relationship, Drug , Gabapentin , Hemodynamics/physiology , In Vitro Techniques , Patch-Clamp Techniques , Potassium Chloride/antagonists & inhibitors , Potassium Chloride/pharmacology , Secretory Vesicles/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL