Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Type of study
Publication year range
1.
Brain Pathol ; : e13279, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38887180

ABSTRACT

Voltage-gated CaV2.1 (P/Q-type) Ca2+ channels play a crucial role in regulating neurotransmitter release, thus contributing to synaptic plasticity and to processes such as learning and memory. Despite their recognized importance in neural function, there is limited information on their potential involvement in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we aimed to explore the impact of AD pathology on the density and nanoscale compartmentalization of CaV2.1 channels in the hippocampus in association with GABAB receptors. Histoblotting experiments showed that the density of CaV2.1 channel was significantly reduced in the hippocampus of APP/PS1 mice in a laminar-dependent manner. CaV2.1 channel was enriched in the active zone of the axon terminals and was present at a very low density over the surface of dendritic tree of the CA1 pyramidal cells, as shown by quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL). In APP/PS1 mice, the density of CaV2.1 channel in the active zone was significantly reduced in the strata radiatum and lacunosum-moleculare, while it remained unaltered in the stratum oriens. The decline in Cav2.1 channel density was found to be associated with a corresponding impairment in the GABAergic synaptic function, as evidenced by electrophysiological experiments carried out in the hippocampus of APP/PS1 mice. Remarkably, double SDS-FRL showed a co-clustering of CaV2.1 channel and GABAB1 receptor in nanodomains (~40-50 nm) in wild type mice, while in APP/PS1 mice this nanoarchitecture was absent. Together, these findings suggest that the AD pathology-induced reduction in CaV2.1 channel density and CaV2.1-GABAB1 de-clustering may play a role in the synaptic transmission alterations shown in the AD hippocampus. Therefore, uncovering these layer-dependent changes in P/Q calcium currents associated with AD pathology can benefit the development of future strategies for AD management.

2.
Mol Psychiatry ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575807

ABSTRACT

Subtype 5 metabotropic glutamate receptors (mGlu5) are known to play an important role in regulating cognitive, social and valence systems. However, it remains largely unknown at which circuits and neuronal types mGlu5 act to influence these behavioral domains. Altered tissue- or cell-specific expression or function of mGlu5 has been proposed to contribute to the exacerbation of neuropsychiatric disorders. Here, we examined how these receptors regulate the activity of somatostatin-expressing (SST+) neurons, as well as their influence on behavior and brain rhythmic activity. Loss of mGlu5 in SST+ neurons elicited excitatory synaptic dysfunction in a region and sex-specific manner together with a range of emotional imbalances including diminished social novelty preference, reduced anxiety-like behavior and decreased freezing during retrieval of fear memories. In addition, the absence of mGlu5 in SST+ neurons during fear processing impaired theta frequency oscillatory activity in the medial prefrontal cortex and ventral hippocampus. These findings reveal a critical role of mGlu5 in controlling SST+ neurons excitability necessary for regulating negative emotional states.

3.
Biomedicines ; 10(4)2022 Mar 23.
Article in English | MEDLINE | ID: mdl-35453499

ABSTRACT

It is without doubt that the gene therapy field is currently in the spotlight for the development of new therapeutics targeting unmet medical needs. Thus, considering the gene therapy scenario, neurological diseases in general and neurodegenerative disorders in particular are emerging as the most appealing choices for new therapeutic arrivals intended to slow down, stop, or even revert the natural progressive course that characterizes most of these devastating neurodegenerative processes. Since an extensive coverage of all available literature is not feasible in practical terms, here emphasis was made in providing some advice to beginners in the field with a narrow focus on elucidating the best delivery route available for fulfilling any given AAV-based therapeutic approach. Furthermore, it is worth nothing that the number of ongoing clinical trials is increasing at a breath-taking speed. Accordingly, a landscape view of preclinical and clinical initiatives is also provided here in an attempt to best illustrate what is ongoing in this quickly expanding field.

4.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203739

ABSTRACT

It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson's disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson's disease and related synucleinopathies.


Subject(s)
Biomedical Research , Dependovirus/genetics , Genetic Vectors/therapeutic use , Parkinson Disease/genetics , Animals , Disease Models, Animal , Gene Transfer Techniques , Humans
5.
J Neurosci ; 38(46): 10002-10015, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30315127

ABSTRACT

The neuromodulatory effects of GABA on pyramidal neurons are mediated by GABAB receptors (GABABRs) that signal via a conserved G-protein-coupled pathway. Two prominent effectors regulated by GABABRs include G-protein inwardly rectifying K+ (GIRK) and P/Q/N type voltage-gated Ca2+ (CaV2) ion channels that control excitability and synaptic output of these neurons, respectively. Regulator of G-protein signaling 7 (RGS7) has been shown to control GABAB effects, yet the specificity of its impacts on effector channels and underlying molecular mechanisms is poorly understood. In this study, we show that hippocampal RGS7 forms two distinct complexes with alternative subunit configuration bound to either membrane protein R7BP (RGS7 binding protein) or orphan receptor GPR158. Quantitative biochemical experiments show that both complexes account for targeting nearly the entire pool of RGS7 to the plasma membrane. We analyzed the effect of genetic elimination in mice of both sexes and overexpression of various components of RGS7 complex by patch-clamp electrophysiology in cultured neurons and brain slices. We report that RGS7 prominently regulates GABABR signaling to CaV2, in addition to its known involvement in modulating GIRK. Strikingly, only complexes containing R7BP, but not GPR158, accelerated the kinetics of both GIRK and CaV2 modulation by GABABRs. In contrast, GPR158 overexpression exerted the opposite effect and inhibited RGS7-assisted temporal modulation of GIRK and CaV2 by GABA. Collectively, our data reveal mechanisms by which distinctly composed macromolecular complexes modulate the activity of key ion channels that mediate the inhibitory effects of GABA on hippocampal CA1 pyramidal neurons.SIGNIFICANCE STATEMENT This study identifies the contributions of distinct macromolecular complexes containing a major G-protein regulator to controlling key ion channel function in hippocampal neurons with implications for understanding molecular mechanisms underlying synaptic plasticity, learning, and memory.


Subject(s)
Caveolin 2/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Hippocampus/physiology , Neurons/physiology , RGS Proteins/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Female , Insecta , Ion Channels/physiology , Male , Mice , Mice, Knockout , Neural Inhibition/physiology
6.
Neuroscience ; 357: 349-362, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28629847

ABSTRACT

The basolateral amygdala receives a very dense cholinergic innervation from the basal forebrain that is important for memory consolidation. Although behavioral studies have shown that both M1 and M2 muscarinic receptors are critical for these mnemonic functions, there have been very few neuroanatomical and electrophysiological investigations of the localization and function of different types of muscarinic receptors in the amygdala. In the present study we investigated the subcellular localization of M2 muscarinic receptors (M2Rs) in the anterior basolateral nucleus (BLa) of the mouse, including the localization of M2Rs in parvalbumin (PV) immunoreactive interneurons, using double-labeling immunoelectron microscopy. Little if any M2R-immunoreactivity (M2R-ir) was observed in neuronal somata, but the neuropil was densely labeled. Ultrastructural analysis using a pre-embedding immunogold-silver technique (IGS) demonstrated M2R-ir in dendritic shafts, spines, and axon terminals forming asymmetrical (excitatory) or symmetrical (mostly inhibitory) synapses. In addition, about one-quarter of PV+ axon terminals and half of PV+ dendrites, localized using immunoperoxidase, were M2R+ when observed in single thin sections. In all M2R+ neuropilar structures, including those that were PV+, about one-quarter to two-thirds of M2R+ immunoparticles were plasma-membrane-associated, depending on the structure. The expression of M2Rs in PV+ and PV-negative terminals forming symmetrical synapses indicates M2R modulation of inhibitory transmission. Electrophysiological studies in mouse and rat brain slices, including paired recordings from interneurons and pyramidal projection neurons, demonstrated M2R-mediated suppression of GABA release. These findings suggest cell-type-specific functions of M2Rs and shed light on organizing principles of cholinergic modulation in the BLa.


Subject(s)
Axons/metabolism , Basolateral Nuclear Complex/metabolism , Dendrites/metabolism , Receptor, Muscarinic M2/metabolism , Synapses/metabolism , Animals , Axons/ultrastructure , Basolateral Nuclear Complex/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dendrites/ultrastructure , Inhibitory Postsynaptic Potentials/physiology , Interneurons/metabolism , Interneurons/ultrastructure , Male , Mice, 129 Strain , Parvalbumins/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats, Sprague-Dawley , Synapses/ultrastructure , Tissue Culture Techniques
7.
Front Neuroanat ; 10: 114, 2016.
Article in English | MEDLINE | ID: mdl-27965545

ABSTRACT

A member of regulator of G-protein signaling family, RGS7, is an essential modulator of signaling through GABAB receptors. RGS7 functions as a macromolecular complex with type 5 G protein ß (Gß5) and R7 binding protein (R7BP) to control the localization and function of the resultant heterotrimeric complexes. Here, we used co-immunoprecipitation, in situ hybridization, histoblot and immunohistochemical techniques at the light and electron microscopic level to advance understanding of RGS7-Gß5-R7BP complexes in the central nervous system, focusing on distinct neuronal populations in the cerebellar cortex. Histoblot analysis showed that RGS7, Gß5 and R7BP proteins were widely expressed in the brain, with mostly an overlapping pattern and showing a high expression level in the molecular layer of the cerebellar cortex. Co-immunoprecipitation experiments established that the RGS7/Gß5 forms complexes with R7BP in the cerebellum. At the cellular level, RGS7 and R7BP mRNAs were expressed at the highest level in Purkinje cells (PCs) and Golgi cells, and at low levels in granule cells. Immunohistochemistry confirmed that labeling for RGS7, Gß5 and R7BP were present in the three neuronal populations and concentrated in dendrites and spines. At the electron microscopic level, immunolabeling for RGS7, Gß5 and R7BP proteins was found both at postsynaptic and presynaptic sites and showed similar distribution patterns. Immunoreactivity for the three proteins was mostly localized along the extrasynaptic plasma membrane of dendritic shafts and spines of PCs and to a lesser extent, in axon terminals (AT) establishing excitatory synapses. Quantitative analysis of immunogold particles for RGS7, Gß5 and R7BP revealed that they are non-uniformly distributed along the surface of PCs, and show enrichment around excitatory synapses on dendritic spines. We further report that deletion of R7BP in mice reduced the targeting of both RGS7 and Gß5 to the plasma membrane. Altogether, these data support the existence of macromolecular complexes composed of RGS7-Gß5-R7BP in PCs. The location at post- and pre-synaptic sites in PCs spines-parallel fiber synapses suggests their involvement in the modulation of glutamatergic neurotransmission in the cerebellar cortex.

8.
J Biol Chem ; 290(22): 13622-39, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25792749

ABSTRACT

Regulators of G protein signaling control the duration and extent of signaling via G protein-coupled receptor (GPCR) pathways by accelerating the GTP hydrolysis on G protein α subunits thereby promoting termination of GPCR signaling. A member of this family, RGS7, plays a critical role in the nervous system where it regulates multiple neurotransmitter GPCRs that mediate vision, memory, and the action of addictive drugs. Previous studies have established that in vivo RGS7 forms mutually exclusive complexes with the membrane protein RGS7-binding protein or the orphan receptor GPR158. In this study, we examine the impact of GPR158 on RGS7 in the brain. We report that knock-out of GPR158 in mice results in marked post-transcriptional destabilization of RGS7 and substantial loss of its association with membranes in several brain regions. We further identified the RGS7-binding site in the C terminus of GPR158 and found that it shares significant homology with the RGS7-binding protein. The proximal portion of the GPR158 C terminus additionally contained a conserved sequence that was capable of enhancing RGS7 GTPase-activating protein activity in solution by an allosteric mechanism acting in conjunction with the regulators of the G protein signaling-binding domain. The distal portion of the GPR158 C terminus contained several phosphodiesterase E γ-like motifs and selectively recruited G proteins in their activated state. The results of this study establish GPR158 as an essential regulator of RGS7 in the native nervous system with a critical role in controlling its expression, membrane localization, and catalytic activity.


Subject(s)
Brain/metabolism , Gene Expression Regulation , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Allosteric Site , Animals , COS Cells , Catalysis , Catalytic Domain , Cloning, Molecular , Computational Biology , Cytosol/metabolism , GTP Phosphohydrolases/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Signal Transduction
9.
Elife ; 3: e02053, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24755289

ABSTRACT

In the hippocampus, the inhibitory neurotransmitter GABA shapes the activity of the output pyramidal neurons and plays important role in cognition. Most of its inhibitory effects are mediated by signaling from GABAB receptor to the G protein-gated Inwardly-rectifying K+ (GIRK) channels. Here, we show that RGS7, in cooperation with its binding partner R7BP, regulates GABABR-GIRK signaling in hippocampal pyramidal neurons. Deletion of RGS7 in mice dramatically sensitizes GIRK responses to GABAB receptor stimulation and markedly slows channel deactivation kinetics. Enhanced activity of this signaling pathway leads to decreased neuronal excitability and selective disruption of inhibitory forms of synaptic plasticity. As a result, mice lacking RGS7 exhibit deficits in learning and memory. We further report that RGS7 is selectively modulated by its membrane anchoring subunit R7BP, which sets the dynamic range of GIRK responses. Together, these results demonstrate a novel role of RGS7 in hippocampal synaptic plasticity and memory formation. DOI: http://dx.doi.org/10.7554/eLife.02053.001.


Subject(s)
CA1 Region, Hippocampal/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/physiology , Memory , Neuronal Plasticity/physiology , RGS Proteins/physiology , Receptors, GABA-B/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Cells, Cultured , Mice , Signal Transduction
10.
Neuron ; 80(1): 159-70, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24094109

ABSTRACT

Repeated cocaine exposure triggers adaptations in layer 5/6 glutamatergic neurons in the medial prefrontal cortex (mPFC) that promote behavioral sensitization and drug-seeking behavior. While suppression of metabotropic inhibitory signaling has been implicated in these behaviors, underlying mechanisms are unknown. Here, we show that Girk/K(IR)3 channels mediate most of the GABA(B) receptor (GABA(B)R)-dependent inhibition of layer 5/6 pyramidal neurons in the mPFC and that repeated cocaine suppresses this pathway. This adaptation was selective for GABA(B)R-dependent Girk signaling in layer 5/6 pyramidal neurons of the prelimbic cortex (PrLC) and involved a D1/5 dopamine receptor- and phosphorylation-dependent internalization of GABA(B)R and Girk channels. Persistent suppression of Girk signaling in layer 5/6 of the dorsal mPFC enhanced cocaine-induced locomotor activity and occluded behavioral sensitization. Thus, the cocaine-induced suppression of GABA(B)R-Girk signaling in layer 5/6 pyramidal neurons of the prelimbic cortex appears to represent an early adaptation critical for promoting addiction-related behavior.


Subject(s)
Cocaine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Signal Transduction/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Behavior, Animal/physiology , Drug-Seeking Behavior/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Receptors, Dopamine D1/metabolism
11.
Hippocampus ; 23(12): 1231-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23804514

ABSTRACT

In the hippocampus, signaling through G protein-coupled receptors is modulated by Regulators of G protein signaling (Rgs) proteins, which act to stimulate the rate of GTP hydrolysis, and consequently, G protein inactivation. The R7-Rgs subfamily selectively deactivates the G(i/o)-class of Gα subunits that mediate the action of several GPCRs. Here, we used co-immunoprecipitation, electrophysiology and immunoelectron microscopy techniques to investigate the formation of macromolecular complexes and spatial relationship of Rgs7/Gß5 complexes and its prototypical signaling partners, the GABAB receptor and Girk channel. Co-expression of recombinant GABAB receptors and Girk channels in combination with co-immunoprecipitation experiments established that the Rgs7/Gß5 forms complexes with GABAB receptors or Girk channels. Using electrophysiological experiments, we found that GABAB -Girk current deactivation kinetics was markedly faster in cells coexpressing Rgs7/Gß5. At the electron microscopic level, immunolabeling for Rgs7 and Gß5 proteins was found primarily in the dendritic layers of the hippocampus and showed similar distribution patterns. Immunoreactivity was mostly localized along the extrasynaptic plasma membrane of dendritic shafts and spines of pyramidal cells and, to a lesser extent, to that of presynaptic terminals. Quantitative analysis of immunogold particles for Rgs7 and Gß5 revealed an enrichment of the two proteins around excitatory synapses on dendritic spines, virtually identical to that of Girk2 and GABAB1 . These data support the existence of macromolecular complexes composed of GABAB receptor-G protein-Rgs7-Girk channels in which Rgs7 and Gß5 proteins may preferentialy modulate GABAB receptor signaling through the deactivation of Girk channels on dendritic spines. In contrast, Rgs7 and Girk2 were associated but mainly segregated from GABAB1 in dendritic shafts, where Rgs7/Gß5 signaling complexes might modulate Girk-dependent signaling via a different metabotropic receptor(s).


Subject(s)
CA1 Region, Hippocampal/cytology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Receptors, GABA/metabolism , Animals , Biophysics , Cell Line, Transformed , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Electric Stimulation , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/ultrastructure , Humans , Imaging, Three-Dimensional , Immunoprecipitation , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Microscopy, Immunoelectron , Neurons/ultrastructure , Patch-Clamp Techniques , RGS Proteins/genetics , RGS Proteins/ultrastructure , Receptors, GABA/genetics , Receptors, GABA/ultrastructure , Transfection
12.
J Neurosci ; 31(34): 12251-7, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865468

ABSTRACT

Enhanced glutamatergic neurotransmission in dopamine (DA) neurons of the ventral tegmental area (VTA), triggered by a single cocaine injection, represents an early adaptation linked to the more enduring effects of abused drugs that characterize addiction. Here, we examined the impact of in vivo cocaine exposure on metabotropic inhibitory signaling involving G-protein-gated inwardly rectifying K(+) (Girk) channels in VTA DA neurons. Somatodendritic Girk currents evoked by the GABA(B) receptor (GABA(B)R) agonist baclofen were diminished in a dose-dependent manner in mice given a single cocaine injection. This adaptation persisted for 3-4 d, was specific for DA neurons of the VTA, and occurred in parallel with an increase in spontaneous glutamatergic neurotransmission. No additional suppression of GABA(B)R-Girk signaling was observed following repeated cocaine administration. While total Girk2 and GABA(B)R1 mRNA and protein levels were unaltered by cocaine exposure in VTA DA neurons, the cocaine-induced decrease in GABA(B)R-Girk signaling correlated with a reduction in Girk2-containing channels at the plasma membrane in VTA DA neurons. Systemic pretreatment with sulpiride, but not SCH23390 (7-chloro-3-methyl-1-phenyl-1,2,4,5-tetrahydro-3-benzazepin-8-ol), prevented the cocaine-induced suppression of GABA(B)R-Girk signaling, implicating D(2/3) DA receptor activation in this adaptation. The acute cocaine-induced weakening of somatodendritic Girk signaling complements the previously demonstrated cocaine-induced strengthening of glutamatergic neurotransmission, likely contributing to enhanced output of VTA DA neurons during the early stages of addiction.


Subject(s)
Cocaine/toxicity , Dopamine/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Neurons/drug effects , Receptors, GABA-B/physiology , Signal Transduction/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Animals , Disease Models, Animal , Dopamine Uptake Inhibitors/pharmacology , Drug Administration Schedule , Female , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Reaction Time/drug effects , Reaction Time/physiology , Signal Transduction/drug effects , Ventral Tegmental Area/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...