Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38313283

ABSTRACT

Opioid receptors within the CNS regulate pain sensation and mood and are key targets for drugs of abuse. Within the adult rodent hippocampus (HPC), µ-opioid receptor agonists suppress inhibitory parvalbumin-expressing interneurons (PV-INs), thus disinhibiting the circuit. However, it is uncertain if this disinhibitory motif is conserved in other cortical regions, species, or across development. We observed that PV-IN mediated inhibition is robustly suppressed by opioids in HPC but not neocortex in mice and nonhuman primates, with spontaneous inhibitory tone in resected human tissue also following a consistent dichotomy. This hippocampal disinhibitory motif was established in early development when immature PV-INs and opioids already influence primordial network rhythmogenesis. Acute opioid-mediated modulation was partially occluded with morphine pretreatment, with implications for the effects of opioids on hippocampal network activity during circuit maturation as well as learning and memory. Together, these findings demonstrate that PV-INs exhibit a divergence in opioid sensitivity across brain regions that is remarkably conserved across evolution and highlights the underappreciated role of opioids acting through immature PV-INs in shaping hippocampal development.

2.
Front Mol Neurosci ; 14: 712609, 2021.
Article in English | MEDLINE | ID: mdl-34630033

ABSTRACT

Medial ganglionic eminence (MGE)-derived parvalbumin (PV)+, somatostatin (SST)+and Neurogliaform (NGFC)-type cortical and hippocampal interneurons, have distinct molecular, anatomical, and physiological properties. However, the molecular mechanisms regulating their maturation remain poorly understood. Here, via single-cell transcriptomics, we show that the obligate NMDA-type glutamate receptor (NMDAR) subunit gene Grin1 mediates transcriptional regulation of gene expression in specific subtypes of MGE-derived interneurons, leading to altered subtype abundances. Notably, MGE-specific early developmental Grin1 loss results in a broad downregulation of diverse transcriptional, synaptogenic and membrane excitability regulatory programs in the juvenile brain. These widespread gene expression abnormalities mirror aberrations that are typically associated with neurodevelopmental disorders. Our study hence provides a road map for the systematic examination of NMDAR signaling in interneuron subtypes, revealing potential MGE-specific genetic targets that could instruct future therapies of psychiatric disorders.

3.
Elife ; 92020 11 05.
Article in English | MEDLINE | ID: mdl-33150866

ABSTRACT

Type I lissencephaly is a neuronal migration disorder caused by haploinsuffiency of the PAFAH1B1 (mouse: Pafah1b1) gene and is characterized by brain malformation, developmental delays, and epilepsy. Here, we investigate the impact of Pafah1b1 mutation on the cellular migration, morphophysiology, microcircuitry, and transcriptomics of mouse hippocampal CA1 parvalbumin-containing inhibitory interneurons (PV+INTs). We find that WT PV+INTs consist of two physiological subtypes (80% fast-spiking (FS), 20% non-fast-spiking (NFS)) and four morphological subtypes. We find that cell-autonomous mutations within interneurons disrupts morphophysiological development of PV+INTs and results in the emergence of a non-canonical 'intermediate spiking (IS)' subset of PV+INTs. We also find that now dominant IS/NFS cells are prone to entering depolarization block, causing them to temporarily lose the ability to initiate action potentials and control network excitation, potentially promoting seizures. Finally, single-cell nuclear RNAsequencing of PV+INTs revealed several misregulated genes related to morphogenesis, cellular excitability, and synapse formation.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , Hippocampus/cytology , Interneurons/metabolism , Microtubule-Associated Proteins/metabolism , Parvalbumins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Electrophysiological Phenomena , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics
4.
Elife ; 92020 06 04.
Article in English | MEDLINE | ID: mdl-32496194

ABSTRACT

The ability to modulate the efficacy of synaptic communication between neurons constitutes an essential property critical for normal brain function. Animal models have proved invaluable in revealing a wealth of diverse cellular mechanisms underlying varied plasticity modes. However, to what extent these processes are mirrored in humans is largely uncharted thus questioning their relevance in human circuit function. In this study, we focus on neurogliaform cells, that possess specialized physiological features enabling them to impart a widespread inhibitory influence on neural activity. We demonstrate that this prominent neuronal subtype, embedded in both mouse and human neural circuits, undergo remarkably similar activity-dependent modulation manifesting as epochs of enhanced intrinsic excitability. In principle, these evolutionary conserved plasticity routes likely tune the extent of neurogliaform cell mediated inhibition thus constituting canonical circuit mechanisms underlying human cognitive processing and behavior.


Subject(s)
Interneurons/physiology , Neuronal Plasticity , Adult , Aged , Animals , Biological Evolution , Brain/physiology , Female , Humans , Interneurons/chemistry , Male , Mice , Middle Aged , Neuroglia/chemistry , Neuroglia/physiology , Pyramidal Cells/chemistry , Pyramidal Cells/physiology , Young Adult
5.
Curr Protoc Neurosci ; 92(1): e93, 2020 06.
Article in English | MEDLINE | ID: mdl-32584517

ABSTRACT

GABAergic interneurons comprise a small but diverse subset of neurons in the mammalian brain that tightly regulate neuronal circuit maturation and information flow and, ultimately, behavior. Because of their centrality in the etiology of numerous neurological disorders, examining the molecular architecture of these neurons under different physiological scenarios has piqued the interest of the broader neuroscience community. The last few years have seen an explosion in next-generation sequencing (NGS) approaches aimed at identifying genetic and state-dependent subtypes in neuronal diversity. Although several approaches are employed to address neuronal molecular diversity, ribosomal tagging has emerged at the forefront of identifying the translatomes of neuronal subtypes. This approach primarily relies on Cre recombinase-driven expression of hemagglutinin A (HA)-tagged RiboTag mice exclusively in the neuronal subtype of interest. This allows the immunoprecipitation of cell-type-specific, ribosome-engaged mRNA, expressed both in the soma and the neuronal processes, for targeted quantitative real-time PCR (qRT-PCR) or high-throughput RNA sequencing analyses. Here we detail the typical technical caveats associated with successful application of the RiboTag technique for analyzing GABAergic interneurons, and in theory other sparse cell types, in the central nervous system. Published 2020. U.S. Government. Basic Protocol 1: Breeding mice to obtain RiboTag homozygosity Support Protocol 1: Detection of ectopic Cre recombinase expression Basic Protocol 2: The RiboTag assay Support Protocol 2: Real-time quantitative PCR (qRT-PCR) assay of RiboTag-derived cell-type-specific RNA Support Protocol 3: Construction of cell-type-specific RNA-seq library Support Protocol 4: Secondary analyses of RiboTag-derived RNA-seq dataset.


Subject(s)
GABAergic Neurons/metabolism , Integrases/metabolism , Interneurons/metabolism , Ribosomes/metabolism , Animals , Female , Immunoprecipitation/methods , Male , Mice, Transgenic , Neurons/metabolism , RNA/metabolism , RNA, Messenger/genetics , Ribosomes/genetics
6.
Front Cell Neurosci ; 14: 36, 2020.
Article in English | MEDLINE | ID: mdl-32161521

ABSTRACT

γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the mature brain but has the paradoxical property of depolarizing neurons during early development. Depolarization provided by GABAA transmission during this early phase regulates neural stem cell proliferation, neural migration, neurite outgrowth, synapse formation, and circuit refinement, making GABA a key factor in neural circuit development. Importantly, depending on the context, depolarizing GABAA transmission can either drive neural activity or inhibit it through shunting inhibition. The varying roles of depolarizing GABAA transmission during development, and its ability to both drive and inhibit neural activity, makes it a difficult developmental cue to study. This is particularly true in the later stages of development when the majority of synapses form and GABAA transmission switches from depolarizing to hyperpolarizing. Here, we addressed the importance of depolarizing but inhibitory (or shunting) GABAA transmission in glutamatergic synapse formation in hippocampal CA1 pyramidal neurons. We first showed that the developmental depolarizing-to-hyperpolarizing switch in GABAA transmission is recapitulated in organotypic hippocampal slice cultures. Based on the expression profile of K+-Cl- co-transporter 2 (KCC2) and changes in the GABA reversal potential, we pinpointed the timing of the switch from depolarizing to hyperpolarizing GABAA transmission in CA1 neurons. We found that blocking depolarizing but shunting GABAA transmission increased excitatory synapse number and strength, indicating that depolarizing GABAA transmission can restrain glutamatergic synapse formation. The increase in glutamatergic synapses was activity-dependent but independent of BDNF signaling. Importantly, the elevated number of synapses was stable for more than a week after GABAA inhibitors were washed out. Together these findings point to the ability of immature GABAergic transmission to restrain glutamatergic synapse formation and suggest an unexpected role for depolarizing GABAA transmission in shaping excitatory connectivity during neural circuit development.

7.
J Cell Sci ; 133(2)2020 01 23.
Article in English | MEDLINE | ID: mdl-31974277

ABSTRACT

Impaired chloride transport affects diverse processes ranging from neuron excitability to water secretion, which underlie epilepsy and cystic fibrosis, respectively. The ability to image chloride fluxes with fluorescent probes has been essential for the investigation of the roles of chloride channels and transporters in health and disease. Therefore, developing effective fluorescent chloride reporters is critical to characterizing chloride transporters and discovering new ones. However, each chloride channel or transporter has a unique functional context that demands a suite of chloride probes with appropriate sensing characteristics. This Review seeks to juxtapose the biology of chloride transport with the chemistries underlying chloride sensors by exploring the various biological roles of chloride and highlighting the insights delivered by studies using chloride reporters. We then delineate the evolution of small-molecule sensors and genetically encoded chloride reporters. Finally, we analyze discussions with chloride biologists to identify the advantages and limitations of sensors in each biological context, as well as to recognize the key design challenges that must be overcome for developing the next generation of chloride sensors.


Subject(s)
Biosensing Techniques/methods , Chlorides/metabolism , Humans
8.
Neuron ; 102(5): 960-975.e6, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31027966

ABSTRACT

Neocortical circuits consist of stereotypical motifs that must self-assemble during development. Recent evidence suggests that the subtype identity of both excitatory projection neurons (PNs) and inhibitory interneurons (INs) is important for this process. We knocked out the transcription factor Satb2 in PNs to induce those of the intratelencephalic (IT) type to adopt a pyramidal tract (PT)-type identity. Loss of IT-type PNs selectively disrupted the lamination and circuit integration of INs derived from the caudal ganglionic eminence (CGE). Strikingly, reprogrammed PNs demonstrated reduced synaptic targeting of CGE-derived INs relative to controls. In control mice, IT-type PNs targeted neighboring CGE INs, while PT-type PNs did not in deep layers, confirming this lineage-dependent motif. Finally, single-cell RNA sequencing revealed that major CGE IN subtypes were conserved after loss of IT PNs, but with differential transcription of synaptic proteins and signaling molecules. Thus, IT-type PNs influence CGE-derived INs in a non-cell-autonomous manner during cortical development.


Subject(s)
Cell Lineage , Interneurons/metabolism , Neocortex/embryology , Synapses/metabolism , Animals , Cell Movement , Gene Expression , Gene Knockout Techniques , Interneurons/cytology , Matrix Attachment Region Binding Proteins/genetics , Mice , Neural Inhibition/physiology , Neural Pathways/embryology , Neurons/cytology , Neurons/metabolism , Pyramidal Tracts/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Telencephalon/cytology , Transcription Factors/genetics
9.
J Physiol ; 597(6): 1677-1690, 2019 03.
Article in English | MEDLINE | ID: mdl-30570751

ABSTRACT

KEY POINTS: Potassium-chloride co-transporter 2 (KCC2) plays a critical role in regulating chloride homeostasis, which is essential for hyperpolarizing inhibition in the mature nervous system. KCC2 interacts with many proteins involved in excitatory neurotransmission, including the GluK2 subunit of the kainate receptor (KAR). We show that activation of KARs hyperpolarizes the reversal potential for GABA (EGABA ) via both ionotropic and metabotropic signalling mechanisms. KCC2 is required for the metabotropic KAR-mediated regulation of EGABA , although ionotropic KAR signalling can hyperpolarize EGABA independent of KCC2 transporter function. The KAR-mediated hyperpolarization of EGABA is absent in the GluK1/2-/- mouse and is independent of zinc release from mossy fibre terminals. The ability of KARs to regulate KCC2 function may have implications in diseases with disrupted excitation: inhibition balance, such as epilepsy, neuropathic pain, autism spectrum disorders and Down's syndrome. ABSTRACT: Potassium-chloride co-transporter 2 (KCC2) plays a critical role in the regulation of chloride (Cl- ) homeostasis within mature neurons. KCC2 is a secondarily active transporter that extrudes Cl- from the neuron, which maintains a low intracellular Cl- concentration [Cl- ]. This results in a hyperpolarized reversal potential of GABA (EGABA ), which is required for fast synaptic inhibition in the mature central nervous system. KCC2 also plays a structural role in dendritic spines and at excitatory synapses, and interacts with 'excitatory' proteins, including the GluK2 subunit of kainate receptors (KARs). KARs are glutamate receptors that display both ionotropic and metabotropic signalling. We show that activating KARs in the hippocampus hyperpolarizes EGABA , thus strengthening inhibition. This hyperpolarization occurs via both ionotropic and metabotropic KAR signalling in the CA3 region, whereas it is absent in the GluK1/2-/- mouse, and is independent of zinc release from mossy fibre terminals. The metabotropic signalling mechanism is dependent on KCC2, although the ionotropic signalling mechanism produces a hyperpolarization of EGABA even in the absence of KCC2 transporter function. These results demonstrate a novel functional interaction between a glutamate receptor and KCC2, a transporter critical for maintaining inhibition, suggesting that the KAR:KCC2 complex may play an important role in excitatory:inhibitory balance in the hippocampus. Additionally, the ability of KARs to regulate chloride homeostasis independently of KCC2 suggests that KAR signalling can regulate inhibition via multiple mechanisms. Activation of kainate-type glutamate receptors could serve as an important mechanism for increasing the strength of inhibition during periods of strong glutamatergic activity.


Subject(s)
Chlorides/metabolism , Inhibitory Postsynaptic Potentials , Pyramidal Cells/metabolism , Receptors, GABA/metabolism , Receptors, Kainic Acid/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiology , Cells, Cultured , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/metabolism , Mossy Fibers, Hippocampal/physiology , Pyramidal Cells/physiology , Symporters/metabolism , K Cl- Cotransporters , GluK2 Kainate Receptor
10.
Proc Natl Acad Sci U S A ; 115(7): E1618-E1626, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29382760

ABSTRACT

Huntington's disease (HD) is classically characterized as a movement disorder, however cognitive impairments precede the motor symptoms by ∼15 y. Based on proteomic and bioinformatic data linking the Huntingtin protein (Htt) and KCC2, which is required for hyperpolarizing GABAergic inhibition, and the important role of inhibition in learning and memory, we hypothesized that aberrant KCC2 function contributes to the hippocampal-associated learning and memory deficits in HD. We discovered that Htt and KCC2 interact in the hippocampi of wild-type and R6/2-HD mice, with a decrease in KCC2 expression in the hippocampus of R6/2 and YAC128 mice. The reduced expression of the Cl--extruding cotransporter KCC2 is accompanied by an increase in the Cl--importing cotransporter NKCC1, which together result in excitatory GABA in the hippocampi of HD mice. NKCC1 inhibition by the FDA-approved NKCC1 inhibitor bumetanide abolished the excitatory action of GABA and rescued the performance of R6/2 mice on hippocampal-associated behavioral tests.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/psychology , Memory Disorders/psychology , Memory , gamma-Aminobutyric Acid/metabolism , Animals , Bumetanide/administration & dosage , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , Male , Memory/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Mice, Transgenic , Solute Carrier Family 12, Member 2/genetics , Solute Carrier Family 12, Member 2/metabolism , Symporters/genetics , Symporters/metabolism , K Cl- Cotransporters
11.
J Neurosci ; 37(45): 10792-10799, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118207

ABSTRACT

Inhibitory circuits are diverse, yet with a poorly understood cell biology. Functional characterization of distinct inhibitory neuron subtypes has not been sufficient to explain how GABAergic neurotransmission sculpts principal cell activity in a relevant fashion. Our Mini-Symposium brings together several emerging mechanisms that modulate GABAergic neurotransmission dynamically from either the presynaptic or the postsynaptic site. The first two talks discuss novel developmental and neuronal subtype-specific contributions to the excitatory/inhibitory balance and circuit maturation. The next three talks examine how interactions between cellular pathways, lateral diffusion of proteins between synapses, and chloride transporter function at excitatory and inhibitory synapses and facilitate inhibitory synapse adaptations. Finally, we address functional differences within GABAergic interneurons to highlight the importance of diverse, flexible, and versatile inputs that shape network function. Together, the selection of topics demonstrates how developmental and activity-dependent mechanisms coordinate inhibition in relation to the excitatory inputs and vice versa.


Subject(s)
Synapses/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/physiology , Animals , Humans , Nerve Net/cytology , Nerve Net/physiology , Neuronal Plasticity
12.
Elife ; 62017 10 13.
Article in English | MEDLINE | ID: mdl-29028184

ABSTRACT

KCC2 is a neuron-specific K+-Cl- cotransporter essential for establishing the Cl- gradient required for hyperpolarizing inhibition in the central nervous system (CNS). KCC2 is highly localized to excitatory synapses where it regulates spine morphogenesis and AMPA receptor confinement. Aberrant KCC2 function contributes to human neurological disorders including epilepsy and neuropathic pain. Using functional proteomics, we identified the KCC2-interactome in the mouse brain to determine KCC2-protein interactions that regulate KCC2 function. Our analysis revealed that KCC2 interacts with diverse proteins, and its most predominant interactors play important roles in postsynaptic receptor recycling. The most abundant KCC2 interactor is a neuronal endocytic regulatory protein termed PACSIN1 (SYNDAPIN1). We verified the PACSIN1-KCC2 interaction biochemically and demonstrated that shRNA knockdown of PACSIN1 in hippocampal neurons increases KCC2 expression and hyperpolarizes the reversal potential for Cl-. Overall, our global native-KCC2 interactome and subsequent characterization revealed PACSIN1 as a novel and potent negative regulator of KCC2.


Subject(s)
Neurons/physiology , Neuropeptides/metabolism , Phosphoproteins/metabolism , Protein Interaction Maps , Symporters/metabolism , Synapses/physiology , Adaptor Proteins, Signal Transducing , Animals , Brain/cytology , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Mass Spectrometry , Mice, Inbred C57BL , Proteomics , K Cl- Cotransporters
13.
Cell Rep ; 20(9): 2156-2168, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854365

ABSTRACT

Although Netos are considered auxiliary subunits critical for kainate receptor (KAR) function, direct evidence for their regulation of native KARs is limited. Because Neto KAR regulation is GluK subunit/Neto isoform specific, such regulation must be determined in cell-type-specific contexts. We demonstrate Neto1/2 expression in somatostatin (SOM)-, cholecystokinin/cannabinoid receptor 1 (CCK/CB1)-, and parvalbumin (PV)-containing interneurons. KAR-mediated excitation of these interneurons is contingent upon Neto1 because kainate yields comparable effects in Neto2 knockouts and wild-types but fails to excite interneurons or recruit inhibition in Neto1 knockouts. In contrast, presynaptic KARs in CCK/CB1 interneurons are dually regulated by both Neto1 and Neto2. Neto association promotes tonic presynaptic KAR activation, dampening CCK/CB1 interneuron output, and loss of this brake in Neto mutants profoundly increases CCK/CB1 interneuron-mediated inhibition. Our results confirm that Neto1 regulates endogenous somatodendritic KARs in diverse interneurons and demonstrate Neto regulation of presynaptic KARs in mature inhibitory presynaptic terminals.


Subject(s)
Dendrites/metabolism , Interneurons/metabolism , LDL-Receptor Related Proteins/metabolism , Membrane Proteins/metabolism , Nerve Net/metabolism , Neural Inhibition , Receptors, Kainic Acid/metabolism , Receptors, Presynaptic/metabolism , Animals , Gamma Rhythm , Ion Channel Gating , Kainic Acid , Mice, Knockout , Mice, Mutant Strains , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate
14.
J Biol Chem ; 292(15): 6190-6201, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28235805

ABSTRACT

Synaptic inhibition depends on a transmembrane gradient of chloride, which is set by the neuron-specific K+-Cl- co-transporter KCC2. Reduced KCC2 levels in the neuronal membrane contribute to the generation of epilepsy, neuropathic pain, and autism spectrum disorders; thus, it is important to characterize the mechanisms regulating KCC2 expression. In the present study, we determined the role of KCC2-protein interactions in regulating total and surface membrane KCC2 expression. Using quantitative immunofluorescence in cultured mouse hippocampal neurons, we discovered that the kainate receptor subunit GluK2 and the auxiliary subunit Neto2 significantly increase the total KCC2 abundance in neurons but that GluK2 exclusively increases the abundance of KCC2 in the surface membrane. Using a live cell imaging assay, we further determined that KCC2 recycling primarily occurs within 1-2 h and that GluK2 produces an ∼40% increase in the amount of KCC2 recycled to the membrane during this time period. This GluK2-mediated increase in surface recycling translated to a significant increase in KCC2 expression in the surface membrane. Moreover, we found that KCC2 recycling is enhanced by protein kinase C-mediated phosphorylation of the GluK2 C-terminal residues Ser-846 and Ser-868. Lastly, using gramicidin-perforated patch clamp recordings, we found that the GluK2-mediated increase in KCC2 recycling to the surface membrane translates to a hyperpolarization of the reversal potential for GABA (EGABA). In conclusion, our results have revealed a mechanism by which kainate receptors regulate KCC2 expression in the hippocampus.


Subject(s)
Cell Membrane/metabolism , Hippocampus/metabolism , Membrane Potentials/physiology , Neurons/metabolism , Receptors, Kainic Acid/metabolism , Symporters/metabolism , Animals , Cell Membrane/genetics , Cells, Cultured , Hippocampus/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neurons/cytology , Receptors, Kainic Acid/genetics , Symporters/genetics , K Cl- Cotransporters , GluK2 Kainate Receptor
15.
J Physiol ; 594(10): 2593-605, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26876607

ABSTRACT

KCC2 is the central regulator of neuronal Cl(-) homeostasis, and is critical for enabling strong hyperpolarizing synaptic inhibition in the mature brain. KCC2 hypofunction results in decreased inhibition and increased network hyperexcitability that underlies numerous disease states including epilepsy, neuropathic pain and neuropsychiatric disorders. The current holy grail of KCC2 biology is to identify how we can rescue KCC2 hypofunction in order to restore physiological levels of synaptic inhibition and neuronal network activity. It is becoming increasingly clear that diverse cellular signals regulate KCC2 surface expression and function including neurotransmitters and neuromodulators. In the present review we explore the existing evidence that G-protein-coupled receptor (GPCR) signalling can regulate KCC2 activity in numerous regions of the nervous system including the hypothalamus, hippocampus and spinal cord. We present key evidence from the literature suggesting that GPCR signalling is a conserved mechanism for regulating chloride homeostasis. This evidence includes: (1) the activation of group 1 metabotropic glutamate receptors and metabotropic Zn(2+) receptors strengthens GABAergic inhibition in CA3 pyramidal neurons through a regulation of KCC2; (2) activation of the 5-hydroxytryptamine type 2A serotonin receptors upregulates KCC2 cell surface expression and function, restores endogenous inhibition in motoneurons, and reduces spasticity in rats; and (3) activation of A3A-type adenosine receptors rescues KCC2 dysfunction and reverses allodynia in a model of neuropathic pain. We propose that GPCR-signals are novel endogenous Cl(-) extrusion enhancers that may regulate KCC2 function.


Subject(s)
Chlorides/physiology , Homeostasis/physiology , Neurons/physiology , Neurotransmitter Agents/physiology , Symporters/physiology , Animals , Humans , Signal Transduction/physiology
16.
Front Cell Neurosci ; 9: 368, 2015.
Article in English | MEDLINE | ID: mdl-26441539

ABSTRACT

Neto2 is a transmembrane protein that interacts with the neuron-specific K(+)-Cl(-) cotransporter (KCC2) in the central nervous system (CNS). Efficient KCC2 transport is essential for setting the neuronal Cl(-) gradient, which is required for fast GABAergic inhibition. Neto2 is required to maintain the normal abundance of KCC2 in neurons, and increases KCC2 function by binding to the active oligomeric form of this cotransporter. In the present study, we characterized GABAergic inhibition and KCC2-mediated neuronal chloride homeostasis in pyramidal neurons from adult hippocampal slices. Using gramicidin perforated patch clamp recordings we found that the reversal potential for GABA (EGABA) was significantly depolarized. We also observed that surface levels of KCC2 and phosphorylation of KCC2 serine 940 (Ser940) were reduced in Neto2(-/-) neurons compared to wild-type controls. To examine GABAergic inhibition we recorded spontaneous inhibitory postsynaptic currents (sIPSCs) and found that Neto2(-/-) neurons had significant reductions in both their amplitude and frequency. Based on the critical role of Neto2 in regulating GABAergic inhibition we rationalized that Neto2-null mice would be prone to seizure activity. We found that Neto2-null mice demonstrated a decrease in the latency to pentylenetetrazole (PTZ)-induced seizures and an increase in seizure severity.

17.
J Neurosci ; 34(45): 14948-60, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25378161

ABSTRACT

Hyperactivity within the ventral hippocampus (vHPC) has been linked to both psychosis in humans and behavioral deficits in animal models of schizophrenia. A local decrease in GABA-mediated inhibition, particularly involving parvalbumin (PV)-expressing GABA neurons, has been proposed as a key mechanism underlying this hyperactive state. However, direct evidence is lacking for a causal role of vHPC GABA neurons in behaviors associated with schizophrenia. Here, we probed the behavioral function of two different but overlapping populations of vHPC GABA neurons that express either PV or GAD65 by selectively inhibiting these neurons with the pharmacogenetic neuromodulator hM4D. We show that acute inhibition of vHPC GABA neurons in adult mice results in behavioral changes relevant to schizophrenia. Inhibiting either PV or GAD65 neurons produced distinct behavioral deficits. Inhibition of PV neurons, affecting ∼80% of the PV neuron population, robustly impaired prepulse inhibition of the acoustic startle reflex (PPI), startle reactivity, and spontaneous alternation, but did not affect locomotor activity. In contrast, inhibiting a heterogeneous population of GAD65 neurons, affecting ∼40% of PV neurons and 65% of cholecystokinin neurons, increased spontaneous and amphetamine-induced locomotor activity and reduced spontaneous alternation, but did not alter PPI. Inhibition of PV or GAD65 neurons also produced distinct changes in network oscillatory activity in the vHPC in vivo. Together, these findings establish a causal role for vHPC GABA neurons in controlling behaviors relevant to schizophrenia and suggest a functional dissociation between the GABAergic mechanisms involved in hippocampal modulation of sensorimotor processes.


Subject(s)
GABAergic Neurons/physiology , Hippocampus/physiology , Interneurons/physiology , Maze Learning , Neural Inhibition , Reflex, Startle , Schizophrenia/physiopathology , Action Potentials , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , GABAergic Neurons/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/drug effects , Interneurons/metabolism , Locomotion , Mice , Parvalbumins/genetics , Parvalbumins/metabolism , Receptor, Muscarinic M4/agonists , Schizophrenia/metabolism , Synaptic Potentials
18.
Neuron ; 83(3): 722-35, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25102562

ABSTRACT

Memories are thought to be sparsely encoded in neuronal networks, but little is known about why a given neuron is recruited or allocated to a particular memory trace. Previous research shows that in the lateral amygdala (LA), neurons with increased CREB are selectively recruited to a fear memory trace. CREB is a ubiquitous transcription factor implicated in many cellular processes. Which process mediates neuronal memory allocation? One hypothesis is that CREB increases neuronal excitability to bias neuronal recruitment, although this has not been shown experimentally. Here we use several methods to increase neuronal excitability and show this both biases recruitment into the memory trace and enhances memory formation. Moreover, artificial activation of these neurons alone is a sufficient retrieval cue for fear memory expression, showing that these neurons are critical components of the memory trace. These results indicate that neuronal memory allocation is based on relative neuronal excitability immediately before training.


Subject(s)
Conditioning, Psychological/physiology , Fear/physiology , Memory/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Amygdala/physiology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Learning , Male , Nervous System Physiological Phenomena , Neurons/metabolism
19.
Cell Rep ; 7(6): 1762-70, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24910435

ABSTRACT

KCC2 is the neuron-specific K+-Cl(-) cotransporter required for maintaining low intracellular Cl(-), which is essential for fast inhibitory synaptic transmission in the mature CNS. Despite the requirement of KCC2 for inhibitory synaptic transmission, understanding of the cellular mechanisms that regulate KCC2 expression and function is rudimentary. We examined KCC2 in its native protein complex in vivo to identify key KCC2-interacting partners that regulate KCC2 function. Using blue native-polyacrylamide gel electrophoresis (BN-PAGE), we determined that native KCC2 exists in a macromolecular complex with kainate-type glutamate receptors (KARs). We found that KAR subunits are required for KCC2 oligomerization and surface expression. In accordance with this finding, acute and chronic genetic deletion of KARs decreased KCC2 function and weakened synaptic inhibition in hippocampal neurons. Our results reveal KARs as regulators of KCC2, significantly advancing our growing understanding of the tight interplay between excitation and inhibition.


Subject(s)
Chlorides/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, Kainic Acid/metabolism , Symporters/metabolism , Animals , Female , Hippocampus/cytology , Homeostasis , Male , Mice, Inbred C57BL , Neurons/cytology , K Cl- Cotransporters
20.
Proc Natl Acad Sci U S A ; 110(9): 3561-6, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401525

ABSTRACT

KCC2 is a neuron-specific K(+)-Cl(-) cotransporter that is essential for Cl(-) homeostasis and fast inhibitory synaptic transmission in the mature CNS. Despite the critical role of KCC2 in neurons, the mechanisms regulating its function are not understood. Here, we show that KCC2 is critically regulated by the single-pass transmembrane protein neuropilin and tolloid like-2 (Neto2). Neto2 is required to maintain the normal abundance of KCC2 and specifically associates with the active oligomeric form of the transporter. Loss of the Neto2:KCC2 interaction reduced KCC2-mediated Cl(-) extrusion, resulting in decreased synaptic inhibition in hippocampal neurons.


Subject(s)
Chlorides/metabolism , Hippocampus/cytology , Membrane Proteins/deficiency , Neurons/metabolism , Symporters/metabolism , Action Potentials/physiology , Amino Acid Sequence , Animals , Biological Transport , Mass Spectrometry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/cytology , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship , Symporters/chemistry , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...