Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Front Mol Neurosci ; 14: 798261, 2021.
Article in English | MEDLINE | ID: mdl-34899186

ABSTRACT

In the central nervous system, the M-current plays a critical role in regulating subthreshold electrical excitability of neurons, determining their firing properties and responsiveness to synaptic input. The M-channel is mainly formed by subunits Kv7.2 and Kv7.3 that co-assemble to form a heterotetrametric channel. Mutations in Kv7.2 and Kv7.3 are associated with hyperexcitability phenotypes including benign familial neonatal epilepsy (BFNE) and neonatal epileptic encephalopathy (NEE). SGK1.1, the neuronal isoform of the serum and glucocorticoids-regulated kinase 1 (SGK1), increases M-current density in neurons, leading to reduced excitability and protection against seizures. Herein, using two-electrode voltage clamp on Xenopus laevis oocytes, we demonstrate that SGK1.1 selectively activates heteromeric Kv7 subunit combinations underlying the M-current. Importantly, activated SGK1.1 increases M-channel activity in the presence of two different epilepsy mutations found in Kv7.2, R207W and A306T. In addition, proximity ligation assays in the N2a cell line allowed us to address the effect of these mutations on Kv7-SGK1.1-Nedd4 molecular associations, a proposed pathway underlying augmentation of M-channel activity by SGK1.1.

2.
Cell Mol Life Sci ; 78(23): 7733-7756, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34709416

ABSTRACT

Dopamine (DA) transporters (DATs) are regulated by trafficking and modulatory processes that probably rely on stable and transient interactions with neighboring proteins and lipids. Using proximity-dependent biotin identification (BioID), we found novel potential partners for DAT, including several membrane proteins, such as the transmembrane chaperone 4F2hc, the proteolipid M6a and a potential membrane receptor for progesterone (PGRMC2). We also detected two cytoplasmic proteins: a component of the Cullin1-dependent ubiquitination machinery termed F-box/LRR-repeat protein 2 (FBXL2), and the enzyme inositol 5-phosphatase 2 (SHIP2). Immunoprecipitation (IP) and immunofluorescence studies confirmed either a physical association or a close spatial proximity between these proteins and DAT. M6a, SHIP2 and the Cullin1 system were shown to increase DAT activity in coexpression experiments, suggesting a functional role for their association. Deeper analysis revealed that M6a, which is enriched in neuronal protrusions (filopodia or dendritic spines), colocalized with DAT in these structures. In addition, the product of SHIP2 enzymatic activity (phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2]) was tightly associated with DAT, as shown by co-IP and by colocalization of mCherry-DAT with a specific biosensor for this phospholipid. PI(3,4)P2 strongly stimulated transport activity in electrophysiological recordings, and conversely, inhibition of SHIP2 reduced DA uptake in several experimental systems including striatal synaptosomes and the dopaminergic cell line SH-SY5Y. In summary, here we report several potential new partners for DAT and a novel regulatory lipid, which may represent new pharmacological targets for DAT, a pivotal protein in dopaminergic function of the brain.


Subject(s)
Brain/metabolism , Cerebral Cortex/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Phospholipids/metabolism , Protein Interaction Domains and Motifs , Receptors, Cell Surface/metabolism , Animals , Biotinylation , Dopamine Plasma Membrane Transport Proteins/chemistry , Phospholipids/chemistry , Rats , Receptors, Cell Surface/chemistry
3.
Neuropharmacology ; 197: 108745, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34375627

ABSTRACT

The voltage-sensitive sodium channel NaV1.1 plays a critical role in regulating excitability of GABAergic neurons and mutations in the corresponding gene are associated to Dravet syndrome and other forms of epilepsy. The activity of this channel is regulated by several protein kinases. To identify novel regulatory kinases we screened a library of activated kinases and we found that AKT1 was able to directly phosphorylate NaV1.1. In vitro kinase assays revealed that the phosphorylation site was located in the C-terminal part of the large intracellular loop connecting domains I and II of NaV1.1, a region that is known to be targeted by other kinases like PKA and PKC. Electrophysiological recordings revealed that activated AKT1 strongly reduced peak Na+ currents and displaced the inactivation curve to more negative potentials in HEK-293 cell stably expressing NaV1.1. These alterations in current amplitude and steady-state inactivation were mimicked by SC79, a specific activator of AKT1, and largely reverted by triciribine, a selective inhibitor. Neurons expressing endogenous NaV1.1 in primary cultures were identified by expressing a fluorescent protein under the NaV1.1 promoter. There, we also observed a strong decrease in the current amplitude after addition of SC79, but small effects on the inactivation parameters. Altogether, we propose a novel mechanism that might regulate the excitability of neural networks in response to AKT1, a kinase that plays a pivotal role under physiological and pathological conditions, including epileptogenesis.


Subject(s)
NAV1.1 Voltage-Gated Sodium Channel/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Electrophysiological Phenomena , Epilepsies, Myoclonic/genetics , HEK293 Cells , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , Nerve Net/drug effects , Neurons/metabolism , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Rats , Ribonucleosides/pharmacology , Sodium Channel Agonists/pharmacology , Sodium Channel Blockers/pharmacology
4.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34453004

ABSTRACT

Postsynaptic N-methyl-D-aspartate receptors (NMDARs) are crucial mediators of synaptic plasticity due to their ability to act as coincidence detectors of presynaptic and postsynaptic neuronal activity. However, NMDARs exist within the molecular context of a variety of postsynaptic signaling proteins, which can fine-tune their function. Here, we describe a form of NMDAR suppression by large-conductance Ca2+- and voltage-gated K+ (BK) channels in the basal dendrites of a subset of barrel cortex layer 5 pyramidal neurons. We show that NMDAR activation increases intracellular Ca2+ in the vicinity of BK channels, thus activating K+ efflux and strong negative feedback inhibition. We further show that neurons exhibiting such NMDAR-BK coupling serve as high-pass filters for incoming synaptic inputs, precluding the induction of spike timing-dependent plasticity. Together, these data suggest that NMDAR-localized BK channels regulate synaptic integration and provide input-specific synaptic diversity to a thalamocortical circuit.


Subject(s)
Excitatory Postsynaptic Potentials , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/metabolism , Somatosensory Cortex/physiology , Synapses/physiology , Animals , Dendrites/physiology , Humans , Ion Transport , Male , Mice , Mice, Inbred C57BL , Neurons/physiology
5.
J Neurosci ; 40(45): 8604-8617, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33046543

ABSTRACT

The second messenger cAMP is an important determinant of synaptic plasticity that is associated with enhanced neurotransmitter release. Long-term potentiation (LTP) at parallel fiber (PF)-Purkinje cell (PC) synapses depends on a Ca2+-induced increase in presynaptic cAMP that is mediated by Ca2+-sensitive adenylyl cyclases. However, the upstream signaling and the downstream targets of cAMP involved in these events remain poorly understood. It is unclear whether cAMP generated by ß-adrenergic receptors (ßARs) is required for PF-PC LTP, although noradrenergic varicosities are apposed in PF-PC contacts. Guanine nucleotide exchange proteins directly activated by cAMP [Epac proteins (Epac 1-2)] are alternative cAMP targets to protein kinase A (PKA) and Epac2 is abundant in the cerebellum. However, whether Epac proteins participate in PF-PC LTP is not known. Immunoelectron microscopy demonstrated that ßARs are expressed in PF boutons. Moreover, activation of these receptors through their agonist isoproterenol potentiated synaptic transmission in cerebellar slices from mice of either sex, an effect that was insensitive to the PKA inhibitors (H-89, KT270) but that was blocked by the Epac inhibitor ESI 05. Interestingly, prior activation of these ßARs occluded PF-PC LTP, while the ß1AR antagonist metoprolol blocked PF-PC LTP, which was also absent in Epac2-/- mice. PF-PC LTP is associated with an increase in the size of the readily releasable pool (RRP) of synaptic vesicles, consistent with the isoproterenol-induced increase in vesicle docking in cerebellar slices. Thus, the ßAR-mediated modulation of the release machinery and the subsequent increase in the size of the RRP contributes to PF-PC LTP.SIGNIFICANCE STATEMENT G-protein-coupled receptors modulate the release machinery, causing long-lasting changes in synaptic transmission that influence synaptic plasticity. Nevertheless, the mechanisms underlying synaptic responses to ß-adrenergic receptor (ßAR) activation remain poorly understood. An increase in the number of synaptic vesicles primed for exocytosis accounts for the potentiation of neurotransmitter release driven by ßARs. This effect is not mediated by the canonical protein kinase A pathway but rather, through direct activation of the guanine nucleotide exchange protein Epac by cAMP. Interestingly, this ßAR signaling via Epac is involved in long term potentiation at cerebellar granule cell-to-Purkinje cell synapses. Thus, the pharmacological activation of ßARs modulates synaptic plasticity and opens therapeutic opportunities to control this phenomenon.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Long-Term Potentiation/physiology , Receptors, Adrenergic, beta/physiology , Synaptic Vesicles/physiology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cerebellum/cytology , Cerebellum/metabolism , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Male , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Purkinje Cells/physiology , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Vesicles/ultrastructure
7.
J Gen Physiol ; 152(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32275741

ABSTRACT

Fragile X mental retardation protein (FMRP) is an RNA-binding protein prominently expressed in neurons. Missense mutations or complete loss of FMRP can potentially lead to fragile X syndrome, a common form of inherited intellectual disability. In addition to RNA regulation, FMRP was also proposed to modulate neuronal function by direct interaction with the large conductance Ca2+- and voltage-activated potassium channel (BK) ß4 regulatory subunits (BKß4). However, the molecular mechanisms underlying FMRP regulation of BK channels were not studied in detail. We have used electrophysiology and super-resolution stochastic optical reconstruction microscopy (STORM) to characterize the effects of FMRP on pore-forming BKα subunits, as well as the association with regulatory subunits BKß4. Our data indicate that, in the absence of coexpressed ß4, FMRP alters the steady-state properties of BKα channels by decreasing channel activation and deactivation rates. Analysis using the Horrigan-Aldrich model revealed alterations in the parameters associated with channel opening (L0) and voltage sensor activation (J0). Interestingly, FMRP also altered the biophysical properties of BKαß4 channels favoring channel opening, although not as dramatically as BKα. STORM experiments revealed clustered multi-protein complexes, consistent with FMRP interacting not only to BKαß4 but also to BKα. Lastly, we found that a partial loss-of-function mutation in FMRP (R138Q) counteracts many of its functional effects on BKα and BKαß4 channels. In summary, our data show that FMRP modulates the function of both BKα and BKαß4 channels.


Subject(s)
Fragile X Mental Retardation Protein , Large-Conductance Calcium-Activated Potassium Channels , Neurons/metabolism , Electrophysiological Phenomena , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism
8.
Neuropharmacology ; 161: 107568, 2019 12 15.
Article in English | MEDLINE | ID: mdl-30885609

ABSTRACT

Dopamine and glutamate transporters (DAT and GLT-1, respectively) share some biophysical characteristics, as both are secondary active carriers coupled to electrochemical ion gradients. In order to identify common or specific components of their respective proteomes, we performed a proximity labelling assay (BioID) in the hippocampal cell line HT22. While most of the identified proteins were specific for each transporter (and will be analyzed elsewhere), we detected two membrane proteins in the shared interactome of GLT-1 and DAT: the transmembrane protein 263 (Tmem263) and the potassium channel protein Kv7.3. However, only Kv7.3 formed immunoprecipitable complexes with GLT-1 and DAT in lysates of transfected HEK293 cells. Moreover, either DAT or GLT-1 co-clustered with Kv7.2/7.3 along the axonal tracts in co-transfected primary neurons, indicating a close spatial proximity between these proteins. Kv7.3, forming heterotetramers with the closely related subunit Kv7.2, underlies the M-currents that control the resting membrane potential and spiking activity in neurons. To investigate whether the presence of the potassium channel affected DAT or GLT-1 function, we performed uptake determinations using radioactive substrate and electrophysiological measurements. Uptake through both transporters was mildly stimulated by the presence of the channel, an effect that was reversed by the potassium channel blocker XE-991. Electrophysiological recording (in transfected HT22 and differentiated SH-SY5Y cells) indicated that the depolarizing effect induced by the presence of the neurotransmitter was reverted by the activity of the potassium channel. Altogether, these data suggest a tight spatial and functional relationship between the DAT/GLT-1 transporters and the Kv7.2/7.3 potassium channel that immediately readjusts the membrane potential of the neuron, probably to limit the neurotransmitter-mediated neuronal depolarization. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Excitatory Amino Acid Transporter 2/metabolism , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Animals , Cell Line , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Electrophysiological Phenomena/genetics , Excitatory Amino Acid Transporter 2/genetics , Female , Glutamic Acid/metabolism , HEK293 Cells , Humans , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Membrane Potentials/genetics , Neurons/metabolism , Potassium Channel Blockers/pharmacology , Pregnancy , Primary Cell Culture , Rats
9.
ACS Chem Neurosci ; 10(4): 2045-2059, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30763065

ABSTRACT

The cycling of synaptic vesicles ensures that neurons can communicate adequately through their synapses on repeated occasions when activity is sustained, and several steps in this cycle are modulated by actin. The effects of pharmacological stabilization of actin with jasplakinolide or its depolymerization with latrunculin A was assessed on the synaptic vesicle cycle at individual boutons of cerebellar granule cells, using FM1-43 imaging to track vesicle recycling and its photoconversion to specifically label recycled organelles. Remarkable differences in the recycling capacity of individual boutons are evident, and their dependence on the actin cytoskeleton for recycling is clear. Disrupting actin dynamics causes a loss of functional boutons, and while this indicates that exo/endocytotic cycling in boutons is fully dependent on such events, this dependence is only partial in other boutons. Indeed, exocytosis and vesicle trafficking are impaired significantly by stabilizing or depolymerizing actin, whereas repositioning recycled vesicles at the active zone seems to be dependent on actin polymerization alone. These findings support the hypothesis that different steps of synaptic vesicle cycling depend on actin dynamics and that such dependence varies among individual boutons.


Subject(s)
Actins/metabolism , Fluorescent Dyes/metabolism , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Actins/analysis , Animals , Cells, Cultured , Depsipeptides/pharmacology , Female , Fluorescent Dyes/analysis , Male , Neurons/chemistry , Neurons/drug effects , Neurons/metabolism , Photochemistry/methods , Pyridinium Compounds/analysis , Quaternary Ammonium Compounds/analysis , Rats , Rats, Wistar , Synapses/chemistry , Synapses/drug effects , Synaptic Vesicles/chemistry , Synaptic Vesicles/drug effects
10.
Neurochem Int ; 123: 125-132, 2019 02.
Article in English | MEDLINE | ID: mdl-29574129

ABSTRACT

GLT-1 is the main glutamate transporter in the brain and its trafficking controls its availability at the cell surface, thereby shaping glutamatergic neurotransmission under physiological and pathological conditions. Extracellular glutamate is known to trigger ubiquitin-dependent GLT-1 internalization from the surface of the cell to the intracellular compartment, yet here we show that internalization also requires the participation of calcium ions. Consistent with previous studies, the addition of glutamate (1 mM) to mixed primary cultures (containing neurons and astrocytes) promotes GLT-1 internalization, an effect that was suppressed in the absence of extracellular Ca2+. The pathways of Ca2+ mobilization by astrocytes were analyzed in these mixed cultures using the genetically encoded calcium sensor GCaMP6f. A complex pattern of calcium entry was activated by glutamate, with a dramatic and rapid rise in the intracellular Ca2+ concentration partially driven by glutamate transporters, especially in the initial stages after exposure to glutamate. The Na+/Ca2+ exchanger (NCX) plays a dominant role in this Ca2+ mobilization and its blockade suppresses the glutamate induced internalization of GLT-1, both in astrocytes and in a more straightforward experimental system like HEK293 cells transiently transfected with GLT-1. This regulatory mechanism might be relevant to control the amount of GLT-1 transporter at the cell surface in conditions like ischemia or traumatic brain injury, where extracellular concentrations of glutamate are persistently elevated and they promote rapid Ca2+ mobilization.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Sodium-Calcium Exchanger/metabolism , Astrocytes/metabolism , Coculture Techniques , Excitatory Amino Acid Transporter 2 , Glutamic Acid/metabolism , HEK293 Cells , Humans , Neurons/metabolism , Protein Transport/physiology , Sodium/metabolism
11.
Glia ; 66(12): 2737-2755, 2018 12.
Article in English | MEDLINE | ID: mdl-30394597

ABSTRACT

We used proximity-dependent biotin identification (BioID) to find proteins that potentially interact with the major glial glutamate transporter, GLT-1, and we studied how these interactions might affect its activity. GTPase Rac1 was one protein identified, and interfering with its GTP/GDP cycle in mixed primary rat brain cultures affected both the clustering of GLT-1 at the astrocytic processes and the transport kinetics, increasing its uptake activity at low micromolar glutamate concentrations in a manner that was dependent on the effector kinase PAK1 and the actin cytoskeleton. Interestingly, the same manipulations had a different effect on another glial glutamate transporter, GLAST, inhibiting its activity. Importantly, glutamate acts through metabotropic receptors to stimulate the activity of Rac1 in astrocytes, supporting the existence of cross-talk between extracellular glutamate and the astrocytic form of the GLT-1 regulated by Rac1. CDC42EP4/BORG4 (a CDC42 effector) was also identified in the BioID screen, and it is a protein that regulates the assembly of septins and actin fibers, influencing the organization of the cytoskeleton. We found that GLT-1 interacts with septins, which reduces its lateral mobility at the cell surface. Finally, the G-protein subunit GNB4 dampens the activity of GLT-1, as revealed by its response to the activator peptide mSIRK, both in heterologous systems and in primary brain cultures. This effect occurs rapidly and thus, it is unlikely to depend on cytoskeletal dynamics. These novel interactions shed new light on the events controlling GLT-1 activity, thereby helping us to better understand how glutamate homeostasis is maintained in the brain.


Subject(s)
Biological Transport/genetics , Excitatory Amino Acid Transporter 2/metabolism , Neuroglia/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Biological Transport/drug effects , Biotinylation , COS Cells , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , Embryo, Mammalian , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Models, Molecular , Neuroglia/drug effects , Neurons/metabolism , Photobleaching , Rats , Transfection
12.
Adv Neurobiol ; 16: 55-83, 2017.
Article in English | MEDLINE | ID: mdl-28828606

ABSTRACT

Glycine plays two roles in neurotransmission. In caudal areas like the spinal cord and the brainstem, it acts as an inhibitory neurotransmitter, but in all regions of the CNS, it also works as a co-agonist with L-glutamate at N-methyl-D-aspartate receptors (NMDARs). The glycine fluxes in the CNS are regulated by two specific transporters for glycine, GlyT1 and GlyT2, perhaps with the cooperation of diverse neutral amino acid transporters like Asc-1 or SNAT5/SN2. While GlyT2 and Asc-1 are neuronal proteins, GlyT1 and SNAT5 are mainly astrocytic, although neuronal forms of GlyT1 also exist. GlyT1 has attracted considerable interest from the medical community and the pharmaceutical industry since compelling evidence indicates a clear association with the functioning of NMDARs, whose activity is decreased in various psychiatric illnesses. By controlling extracellular glycine, transporter inhibitors might potentiate the activity of NMDARs without activating excitotoxic processes. Physiologically, GlyT1 is a central actor in the cross talk between glutamatergic, glycinergic, dopaminergic, and probably other neurotransmitter systems. Many of these relationships begin to be unraveled by studies performed in recent years using genetic and pharmacological models. These studies are also clarifying the interactions between glycine, glycine transporters, and other co-agonists of the glycine site of NMDARs like D-serine. These findings are also relevant to understand the pathophysiology of devastating diseases like schizophrenia, depression, anxiety, epilepsy, stroke, and chronic pain.


Subject(s)
Brain/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Animals , Glycine/metabolism , Humans , Mice
13.
Neuropharmacology ; 125: 99-116, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28734869

ABSTRACT

Glycinergic inhibitory neurons of the spinal dorsal horn exert critical control over the conduction of nociceptive signals to higher brain areas. The neuronal glycine transporter 2 (GlyT2) is involved in the recycling of synaptic glycine from the inhibitory synaptic cleft and its activity modulates intra and extracellular glycine concentrations. In this report we show that the stimulation of P2X purinergic receptors with ßγ-methylene adenosine 5'-triphosphate induces the up-regulation of GlyT2 transport activity by increasing total and plasma membrane expression and reducing transporter ubiquitination. We identified the receptor subtypes involved by combining pharmacological approaches, siRNA-mediated protein knockdown, and dorsal root ganglion cell enrichment in brainstem and spinal cord primary cultures. Up-regulation of GlyT2 required the combined stimulation of homomeric P2X3 and P2X2 receptors or heteromeric P2X2/3 receptors. We measured the spontaneous glycinergic currents, glycine release and GlyT2 uptake concurrently in response to P2X receptor agonists, and showed that the impact of P2X3 receptor activation on glycinergic neurotransmission involves the modulation of GlyT2 expression or activity. The recognized pro-nociceptive action of P2X3 receptors suggests that the fine-tuning of GlyT2 activity may have consequences in nociceptive signal conduction.


Subject(s)
Cell Membrane/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Cell Membrane/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Glycine/metabolism , Neurons/drug effects , Neurons/metabolism , Pain/metabolism , Purinergic P2X Receptor Agonists/pharmacology , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques , Ubiquitination/drug effects , Ubiquitination/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
14.
J Neurochem ; 142(3): 350-364, 2017 08.
Article in English | MEDLINE | ID: mdl-28445587

ABSTRACT

Cannabinoid receptors mediate short-term retrograde inhibition of neurotransmitter release, as well as long-term depression of synaptic transmission at excitatory synapses. The responses of individual nerve terminals in VGLUT1-pHluorin transfected cerebellar granule cells to cannabinoids have shown that prolonged activation of cannabinoid type 1 receptors (CB1Rs) silences a subpopulation of previously active synaptic boutons. Adopting a combined pharmacological and genetic approach to study the molecular mechanisms of CB1R-induced silencing, we found that adenylyl cyclase inhibition decreases cAMP levels while it increases the number of silent synaptic boutons and occludes the induction of further silencing by the cannabinoid agonist HU-210. Guanine nucleotide exchange proteins directly activated by cAMP (Epac proteins) mediate some of the presynaptic effects of cAMP in the potentiation of synaptic transmission. ESI05, a selective Epac2 inhibitor, and U-73122, the specific inhibitor of phospholipase C (PLC), both augment the number of silent synaptic boutons. Moreover, they abolish the capacity of the Epac activator, 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate monosodium hydrate, to prevent HU-210-induced silencing consistent with PLC signaling lying downstream of Epac2 proteins. Furthermore, Rab3-interacting molecule (RIM)1α KO cells have many more basally silent synaptic boutons (12.9 ± 3.5%) than wild-type cells (1.1 ± 0.5%). HU-210 induced further silencing in these mutant cells, although 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate monosodium hydrate only awoke the HU-210-induced silence and not the basally silent synaptic boutons. This behavior can be rescued by expressing RIM1α in RIM1α KO cells, these cells behaving very much like wild-type cells. These findings support the hypothesis that a cAMP/Epac/PLC signaling pathway targeting the release machinery appears to mediate cannabinoid-induced presynaptic silencing.


Subject(s)
Cerebellum/cytology , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission/drug effects , Animals , Cerebellum/drug effects , Cyclic AMP/metabolism , Down-Regulation/drug effects , Estrenes/pharmacology , Female , Guanine Nucleotide Exchange Factors/metabolism , Male , Neurons/drug effects , Pyrrolidinones/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/drug effects , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/physiology , Type C Phospholipases/metabolism
15.
Neuropharmacology ; 101: 412-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26211974

ABSTRACT

The co-existence of presynaptic G protein coupled receptors, GPCRs, has received little attention, despite the fact that interplay between the signaling pathways activated by such receptors may affect the neurotransmitter release. Using immunocytochemistry and immuhistochemistry we show that mGlu7 and ß-adrenergic receptors are co-expressed in a sub-population of cerebrocortical nerve terminals. mGlu7 receptors readily couple to pathways that inhibit glutamate release. We found that when mGlu7 receptors are also coupled to pathways that enhance glutamate release by prolonged exposure to agonist, and ß-adrenergic receptors are also activated, a cross-talk between their signaling pathways occurs that affect the overall release response. This interaction is the result of mGlu7 receptors inhibiting the adenylyl cyclase activated by ß adrenergic receptors. Thus, blocking Gi/o proteins with pertussis toxin provokes a further increase in release after receptor co-activation which is also observed after activating ß-adrenergic receptor signaling pathways downstream of adenylyl cyclase with the cAMP analog Sp8Br or 8pCPT-2-OMe-cAMP (a specific activator of the guanine nucleotide exchange protein directly activated by cAMP, EPAC). Co-activation of mGlu7 and ß-adrenergic receptors also enhances PLC-dependent accumulation of IP1 and the translocation of the active zone protein Munc13-1 to the membrane, indicating that release potentiation by these receptors involves the modulation of the release machinery.


Subject(s)
Nerve Endings/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Calcium Ionophores/pharmacology , Cerebellum/cytology , Cerebral Cortex/cytology , Cyclic AMP/metabolism , Glutamic Acid/metabolism , Inositol Phosphates/pharmacology , Ionomycin/pharmacology , Mice , Mice, Inbred C57BL , Nerve Endings/drug effects , Nerve Tissue Proteins/metabolism , Propionates/pharmacology , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptophysin/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Thionucleotides/pharmacology
16.
Proc Natl Acad Sci U S A ; 112(7): 2239-44, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646456

ABSTRACT

We describe a fast activity-dependent homeostatic regulation of intrinsic excitability of identified neurons in mouse dorsal striatum, the striatal output neurons. It can be induced by brief bursts of activity, is expressed on a time scale of seconds, limits repetitive firing, and can convert regular firing patterns to irregular ones. We show it is due to progressive recruitment of the KCNQ2/3 channels that generate the M current. This homeostatic mechanism is significantly reduced in striatal output neurons of the R6/2 transgenic mouse model of Huntington's disease, at an age when the neurons are hyperactive in vivo and the mice begin to exhibit locomotor impairment. Furthermore, it can be rescued by bath perfusion with retigabine, a KCNQ channel activator, and chronic treatment improves locomotor performance. Thus, M-current dysfunction may contribute to the hyperactivity and network dysregulation characteristic of this neurodegenerative disease, and KCNQ2/3 channel regulation may be a target for therapeutic intervention.


Subject(s)
Corpus Striatum/physiopathology , Disease Models, Animal , Homeostasis , Huntington Disease/physiopathology , Locomotion , Animals , Mice
17.
PLoS One ; 9(2): e88594, 2014.
Article in English | MEDLINE | ID: mdl-24533119

ABSTRACT

Cannabinoid receptors are the most abundant G protein-coupled receptors in the brain and they mediate retrograde short-term inhibition of neurotransmitter release, as well as long-term depression of synaptic transmission at many excitatory synapses. The induction of presynaptically silent synapses is a means of modulating synaptic strength, which is important for synaptic plasticity. Persistent activation of cannabinoid type 1 receptors (CB1Rs) mutes GABAergic terminals, although it is unclear if CB1Rs can also induce silencing at glutamatergic synapses. Cerebellar granule cells were transfected with VGLUT1-pHluorin to visualise the exo-endocytotic cycle. We found that prolonged stimulation (10 min) of cannabinoid receptors with the agonist HU-210 induces the silencing of previously active synapses. However, the presynaptic silencing induced by HU-210 is transient as it reverses after 20 min. cAMP with forskolin prevented CB1R-induced synaptic silencing, via activation of the Exchange Protein directly Activated by cAMP (Epac). Furthermore, Epac activation accelerated awakening of already silent boutons. Electron microscopy revealed that silencing was associated with synaptic vesicle (SV) redistribution within the nerve terminal, which diminished the number of vesicles close to the active zone of the plasma membrane. Finally, by combining functional and immunocytochemical approaches, we observed a strong correlation between the release capacity of the nerve terminals and RIM1α protein content, but not that of Munc13-1 protein. These results suggest that prolonged stimulation of cannabinoid receptors can transiently silence glutamatergic nerve terminals.


Subject(s)
Cerebellum/cytology , Neurons/physiology , Receptor, Cannabinoid, CB1/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Dronabinol/analogs & derivatives , Dronabinol/chemistry , Endocytosis , Exocytosis , Female , Fluorescent Dyes/chemistry , Male , Nerve Tissue Proteins/metabolism , Pyridinium Compounds/chemistry , Quaternary Ammonium Compounds/chemistry , Rats , Rats, Wistar , Time Factors
18.
BMC Neurosci ; 14: 127, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24138605

ABSTRACT

BACKGROUND: In terms of vesicular recycling, synaptic efficiency is a key determinant of the fidelity of synaptic transmission. The ability of a presynaptic terminal to reuse its vesicular content is thought to be a signature of synaptic maturity and this process depends on the activity of several proteins that govern exo/endocytosis. Upon stimulation, individual terminals in networks of cultured cerebellar granule neurons exhibit heterogeneous exocytic responses, which reflect the distinct states of maturity and plasticity intrinsic to individual synaptic terminals. This dynamic scenario serves as the substrate for processes such as scaling, plasticity and synaptic weight redistribution. Presynaptic strength has been associated with the activity of several types of proteins, including the scaffolding proteins that form the active zone cytomatrix and the proteins involved in presynaptic exocytosis. METHODS: We have combined fluorescence imaging techniques using the styryl dye FM1-43 in primary cultures of cerebellar granule cells with subsequent post-hoc immunocytochemistry in order to study synaptic efficiency in terms of vesicular release. We describe a protocol to easily quantify these results with minimal user intervention. RESULTS: In this study we describe a technique that specifically correlates presynaptic activity with the levels of presynaptic markers. This method involves the use of the styryl dye FM1-43 to estimate the release capacity of a synaptic terminal, and the subsequent post-hoc immunolabelling of thousands of individual nerve terminals. We observed a strong correlation between the release capacity of the nerve terminal and the levels of the RIM1α but not the Munc13-1 protein in the active zone. CONCLUSIONS: Our findings support those of previous studies and point out to RIM1α as a crucial factor in determining synaptic efficiency. These results also demonstrate that this technique is a useful tool to analyse the molecular differences underlying the heterogeneous responses exhibited by neuronal networks.


Subject(s)
Immunohistochemistry/methods , Neurons/physiology , Optical Imaging/methods , Synaptic Transmission/physiology , Animals , Cerebellum/physiology , Fluorescent Dyes , Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Pyridinium Compounds , Quaternary Ammonium Compounds
19.
J Biol Chem ; 288(43): 31370-85, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24036110

ABSTRACT

The adenylyl cyclase activator forskolin facilitates synaptic transmission presynaptically via cAMP-dependent protein kinase (PKA). In addition, cAMP also increases glutamate release via PKA-independent mechanisms, although the downstream presynaptic targets remain largely unknown. Here, we describe the isolation of a PKA-independent component of glutamate release in cerebrocortical nerve terminals after blocking Na(+) channels with tetrodotoxin. We found that 8-pCPT-2'-O-Me-cAMP, a specific activator of the exchange protein directly activated by cAMP (Epac), mimicked and occluded forskolin-induced potentiation of glutamate release. This Epac-mediated increase in glutamate release was dependent on phospholipase C, and it increased the hydrolysis of phosphatidylinositol 4,5-bisphosphate. Moreover, the potentiation of glutamate release by Epac was independent of protein kinase C, although it was attenuated by the diacylglycerol-binding site antagonist calphostin C. Epac activation translocated the active zone protein Munc13-1 from soluble to particulate fractions; it increased the association between Rab3A and RIM1α and redistributed synaptic vesicles closer to the presynaptic membrane. Furthermore, these responses were mimicked by the ß-adrenergic receptor (ßAR) agonist isoproterenol, consistent with the immunoelectron microscopy and immunocytochemical data demonstrating presynaptic expression of ßARs in a subset of glutamatergic synapses in the cerebral cortex. Based on these findings, we conclude that ßARs couple to a cAMP/Epac/PLC/Munc13/Rab3/RIM-dependent pathway to enhance glutamate release at cerebrocortical nerve terminals.


Subject(s)
Cerebral Cortex/metabolism , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Glutamic Acid/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Adrenergic, beta/metabolism , rab3A GTP-Binding Protein/metabolism , Adjuvants, Immunologic/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cerebral Cortex/cytology , Colforsin/pharmacology , Enzyme Inhibitors/pharmacology , Isoproterenol/pharmacology , Mice , Naphthalenes/pharmacology , Presynaptic Terminals/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Synaptic Membranes/metabolism , Synaptic Transmission/physiology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
20.
Neuropharmacology ; 67: 213-22, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23174341

ABSTRACT

Calcium channels that mediate glutamate release (N-type and P/Q-type) are expressed in distinct populations of cerebrocortical nerve terminals in adult mice. mGlu7 receptors are exclusively expressed in nerve terminals containing N-type Ca(2+) channels, which are less tightly coupled to glutamate release than P/Q-type Ca(2+) channels. We recently reported that in addition to inhibit, mGlu7 receptors can also potentiate glutamate release via phosphatidyl inositol (4,5)-bisphosphate hydrolysis and activation of the non-kinase diacylglycerol binding protein Munc13-1, a protein that primes synaptic vesicles for exocytosis. Here, we assessed whether mGlu7 receptor-mediated potentiation of glutamate release is restricted to nerve terminals expressing N-type Ca(2+) channels to compensate for their weak coupling to release. In the hippocampus, mGlu7 receptors are expressed both in nerve terminals containing N-type Ca(2+) channels and in nerve terminals containing P/Q-type Ca(2+) channels. When analyzed, we observed potentiation of mGlu7 receptor mediated release in wild type hippocampal nerve terminals at physiological (1.3 mM) and low (0.1 mM) concentrations of external Ca(2+). By contrast, in nerve terminals from mice lacking the α1B subunit of N-type channels (Ca(v)2.2), in which evoked release is mediated by P/Q-type channels only, no release potentiation was observed at 1.3 mM Ca(2+). We conclude that release potentiation at 1.3 mM [Ca(2+)](e) occurs in nerve terminals expressing N-type channels, whereas that which occurs at low 0.1 mM [Ca(2+)](e) represents the release from nerve terminals containing P/Q-type Ca(2+) channels. Although, mGlu7 receptor mediated potentiation is independent of Ca(2+) channel activity, as it was induced by the Ca(2+) ionophore ionomycin, release potentiation is influenced by the Ca(2+) channel type and/or the associated release machinery.


Subject(s)
Calcium Channels, N-Type/physiology , Calcium Channels, P-Type/physiology , Calcium Channels, Q-Type/physiology , Glutamic Acid/metabolism , Nerve Endings/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Exocytosis/physiology , Female , Male , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL