Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neurosci ; 42(1): 44-57, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34759028

ABSTRACT

The primary somatosensory cortex (S1) is important for the control of movement as it encodes sensory input from the body periphery and external environment during ongoing movement. Mouse S1 consists of several distinct sensorimotor subnetworks that receive topographically organized corticocortical inputs from distant sensorimotor areas, including the secondary somatosensory cortex (S2) and primary motor cortex (M1). The role of the vibrissal S1 area and associated cortical connections during active sensing is well documented, but whether (and if so, how) non-whisker S1 areas are involved in movement control remains relatively unexplored. Here, we demonstrate that unilateral silencing of the non-whisker S1 area in both male and female mice disrupts hind paw movement during locomotion on a rotarod and a runway. S2 and M1 provide major long-range inputs to this S1 area. Silencing S2→non-whisker S1 projections alters the hind paw orientation during locomotion, whereas manipulation of the M1 projection has little effect. Using patch-clamp recordings in brain slices from male and female mice, we show that S2 projection preferentially innervates inhibitory interneuron subtypes. We conclude that interneuron-mediated S2-S1 corticocortical interactions are critical for efficient locomotion.SIGNIFICANCE STATEMENT Somatosensory cortex participates in controlling rhythmic movements, such as whisking and walking, but the neural circuitry underlying movement control by somatosensory cortex remains relatively unexplored. We uncover a corticocortical circuit in primary somatosensory cortex that regulates paw orientation during locomotion in mice. We identify neuronal elements that comprise these cortical pathways using pharmacology, behavioral assays, and circuit-mapping methods.


Subject(s)
Efferent Pathways/physiology , Interneurons/physiology , Orientation, Spatial/physiology , Somatosensory Cortex/physiology , Animals , Female , Locomotion/physiology , Male , Mice , Movement/physiology
2.
J Neurosci ; 41(11): 2360-2372, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33514676

ABSTRACT

Human fMRI studies show that insulin influences brain activity in regions that mediate reward and motivation, including the nucleus accumbens (NAc). Insulin receptors are expressed by NAc medium spiny neurons (MSNs), and studies of cultured cortical and hippocampal neurons suggest that insulin influences excitatory transmission via presynaptic and postsynaptic mechanisms. However, nothing is known about how insulin influences excitatory transmission in the NAc. Furthermore, insulin dysregulation accompanying obesity is linked to cognitive decline, depression, anxiety, and altered motivation that rely on NAc excitatory transmission. Using whole-cell patch-clamp and biochemical approaches, we determined how insulin affects NAc glutamatergic transmission in nonobese and obese male rats and the underlying mechanisms. We find that there are concentration-dependent, bidirectional effects of insulin on excitatory transmission, with insulin receptor activation increasing and IGF receptor activation decreasing NAc excitatory transmission. Increases in excitatory transmission were mediated by activation of postsynaptic insulin receptors located on MSNs. However, this effect was due to an increase in presynaptic glutamate release. This suggested feedback from MSNs to presynaptic terminals. In additional experiments, we found that insulin-induced increases in presynaptic glutamate release are mediated by opioid receptor-dependent disinhibition. Furthermore, obesity resulted in a loss of insulin receptor-mediated increases in excitatory transmission and a reduction in NAc insulin receptor surface expression, while preserving reductions in transmission mediated by IGF receptors. These results provide the first insights into how insulin influences excitatory transmission in the adult brain, and evidence for a previously unidentified form of opioid receptor-dependent disinhibition of NAc glutamatergic transmission.SIGNIFICANCE STATEMENT Data here provide the first insights into how insulin influences excitatory transmission in the adult brain, and identify previously unknown interactions between insulin receptor activation, opioids, and glutamatergic transmission. These data contribute to our fundamental understanding of insulin's influence on brain motivational systems and have implications for the use of insulin as a cognitive enhancer and for targeting of insulin receptors and IGF receptors to alter motivation.


Subject(s)
Endorphins/pharmacology , Glutamic Acid/metabolism , Insulin/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptor, Insulin/drug effects , Synaptic Transmission/drug effects , Animals , Diet, High-Fat , Male , Neurons/drug effects , Obesity/genetics , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/agonists , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D2/drug effects
3.
J Neurophysiol ; 122(3): 1264-1273, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31365322

ABSTRACT

The nucleus accumbens (NAc) plays critical roles in motivated behaviors, including food seeking and feeding. Differences in NAc function contribute to overeating that drives obesity, but the underlying mechanisms are poorly understood. In addition, there is a fair degree of variation in individual susceptibility versus resistance to obesity that is due in part to differences in NAc function. For example, using selectively bred obesity-prone and obesity-resistant rats, we have found that excitability of medium spiny neurons (MSNs) within the NAc core is enhanced in obesity-prone versus -resistant populations, before any diet manipulation. However, it is unknown whether consumption of sugary, fatty "junk food" alters MSN excitability. Here whole cell patch-clamp recordings were conducted to examine MSN intrinsic excitability in adult male obesity-prone and obesity-resistant rats with and without exposure to a sugary, fatty junk food diet. We replicated our initial finding that basal excitability is enhanced in obesity-prone versus obesity-resistant rats and determined that this is due to a lower fast transient potassium current (IA) in prone versus resistant groups. In addition, the junk food diet had opposite effects on excitability in obesity-prone versus obesity-resistant rats. Specifically, junk food enhanced excitability in MSNs of obesity-resistant rats; this was mediated by a reduction in IA. In contrast, junk food reduced excitability in MSNs from obesity-prone rats; this was mediated by an increase in inward-rectifying potassium current. Thus individual differences in obesity susceptibility influence both basal excitability and how MSN excitability adapts to junk food consumption.NEW & NOTEWORTHY Medium spiny neurons (MSNs) in the nucleus accumbens of obesity-prone rats are hyperexcitable compared with MSNs from obesity-resistant rats. We found that 10 days of "junk food" exposure reduces MSN excitability in obesity-prone rats by increasing inward-rectifying potassium current and increases MSN excitability in obesity-resistant rats by decreasing fast transient potassium current. These data show that there are basal and junk food diet-induced differences in MSN excitability in obesity-prone and obesity-resistant individuals; this may contribute to previously observed differences in incentive motivation.


Subject(s)
Action Potentials/physiology , Diet, Carbohydrate Loading , Diet, High-Fat , Disease Susceptibility/physiopathology , Nucleus Accumbens/physiology , Obesity/physiopathology , Potassium Channels, Voltage-Gated/physiology , Animals , Male , Patch-Clamp Techniques , Rats
4.
Neuropharmacology ; 141: 296-304, 2018 10.
Article in English | MEDLINE | ID: mdl-30189184

ABSTRACT

A single base mutation in the Gαi2 protein (G184S) renders this Gα subunit insensitive to the negative modulatory effects of Regulator of G-protein Signaling (RGS) proteins. Mice expressing this RGS insensitive (RGSi) variant of Gαi2 (RGSi Gαi2) display a spontaneous antidepressant-like phenotype that is reversible by treatment with the 5-HT1A receptor (5-HT1AR) antagonist WAY100635. Here we test the hypothesis that increased activity of 5-HT1ARs in the hippocampus of RGSi Gαi2 knock-in mice is responsible for the expression of the observed antidepressant-like behavior. We administered the 5-HT1AR antagonist WAY100635 or the agonist 8-OH-DPAT via bilateral intra-hippocampal infusion cannulae and evaluated antidepressant-like behavior using the tail suspension test (TST). WAY100635 reversed the antidepressant-like phenotype of the RGSi Gαi2 knock-in mice and 8-OH-DPAT produced an antidepressant-like response in wild type mice that was blocked by systemic WAY100635. Furthermore, intra-hippocampal infusion of the RGS19/4 inhibitor CCG-203769 produced an antidepressant-like effect in female mice. Ex-vivo slice recording confirmed the 5-HT1AR-mediated decrease in hippocampal CA1 pyramidal neuron excitability was enhanced in the RGSi Gαi2 knock-in mice. There was no change in hippocampal 5-HT1AR expression as measured by ligand binding but there was a compensatory reduction in Gαi proteins. The findings demonstrate that RGS protein control of hippocampal 5-HT1AR signaling is necessary and sufficient to account for the antidepressant-like phenotype in the RGSi Gαi2 knock-in mice and that RGS proteins highly expressed in the hippocampus should be investigated as targets for novel antidepressant therapies.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/metabolism , Hippocampus/metabolism , Immobility Response, Tonic/physiology , RGS Proteins/antagonists & inhibitors , Receptor, Serotonin, 5-HT1A/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/antagonists & inhibitors , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Female , GTP-Binding Protein alpha Subunit, Gi2/genetics , Gene Knock-In Techniques , Hippocampus/drug effects , Male , Mice , Microinjections , Phenotype , Piperazines/pharmacology , Pyramidal Cells/physiology , Pyridines/pharmacology , Receptor, Serotonin, 5-HT1A/biosynthesis , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology
5.
J Pharmacol Exp Ther ; 363(2): 148-155, 2017 11.
Article in English | MEDLINE | ID: mdl-28838956

ABSTRACT

Neuroadaptive responses to chronic ethanol, such as behavioral sensitization, are associated with N-methyl-D-aspartate receptor (NMDAR) recruitment. Ethanol enhances GluN2B-containing NMDAR function and phosphorylation (Tyr-1472) of the GluN2B-NMDAR subunit in the dorsal medial striatum (DMS) through a protein kinase A (PKA)-dependent pathway. Ethanol-induced phosphorylation of PKA substrates is partially mediated by calcium-stimulated adenylyl cyclase 1 (AC1), which is enriched in the dorsal striatum. As such, AC1 is poised as an upstream modulator of ethanol-induced DMS neuroadaptations that promote drug responding, and thus represents a therapeutic target. Our hypothesis is that loss of AC1 activity will prevent ethanol-induced locomotor sensitization and associated DMS GluN2B-NMDAR adaptations. We evaluated AC1's contribution to ethanol-evoked locomotor responses and DMS GluN2B-NMDAR phosphorylation and function using AC1 knockout (AC1KO) mice. Results were mechanistically validated with the AC1 inhibitor, NB001. Acute ethanol (2.0 g/kg) locomotor responses in AC1KO and wild-type (WT) mice pretreated with NB001 (10 mg/kg) were comparable to WT ethanol controls. However, repeated ethanol treatment (10 days, 2.5 g/kg) failed to produce sensitization in AC1KO or NB001 pretreated mice, as observed in WT ethanol controls, following challenge exposure (2.0 g/kg). Repeated exposure to ethanol in the sensitization procedure significantly increased pTyr-1472 GluN2B levels and GluN2B-containing NMDAR transmission in the DMS of WT mice. Loss of AC1 signaling impaired ethanol-induced increases in DMS pGluN2B levels and NMDAR-mediated transmission. Together, these data support a critical and specific role for AC1 in striatal signaling that mediates ethanol-induced behavioral sensitization, and identify GluN2B-containing NMDARs as an important AC1 target.


Subject(s)
Adenylyl Cyclases/deficiency , Corpus Striatum/metabolism , Ethanol/administration & dosage , Locomotion/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Corpus Striatum/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology
6.
Physiol Rep ; 5(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-28108647

ABSTRACT

Rett syndrome (RTT) is mostly caused by mutations of the X-linked MECP2 gene. Although the causal neuronal mechanisms are still unclear, accumulating experimental evidence obtained from Mecp2-/Y mice suggests that imbalanced excitation/inhibition in central neurons plays a major role. Several approaches may help to rebalance the excitation/inhibition, including agonists of GABAA receptors (GABAAR). Indeed, our previous studies have shown that early-life exposure of Mecp2-null mice to the extrasynaptic GABAAR agonist THIP alleviates several RTT-like symptoms including breathing disorders, motor dysfunction, social behaviors, and lifespan. However, how the chronic THIP affects the Mecp2-/Y mice at the cellular level remains elusive. Here, we show that the THIP exposure in early lives markedly alleviated hyperexcitability of two types of brainstem neurons in Mecp2-/Y mice. In neurons of the locus coeruleus (LC), known to be involved in breathing regulation, the hyperexcitability showed clear age-dependence, which was associated with age-dependent deterioration of the RTT-like breathing irregularities. Both the neuronal hyperexcitability and the breathing disorders were relieved with early THIP treatment. In neurons of the mesencephalic trigeminal nucleus (Me5), both the neuronal hyperexcitability and the changes in intrinsic membrane properties were alleviated with the THIP treatment in Mecp2-null mice. The effects of THIP on both LC and Me5 neuronal excitability remained 1 week after withdrawal. Persistent alleviation of breathing abnormalities in Mecp2-/Y mice was also observed a week after THIP withdrawal. These results suggest that early-life exposure to THIP, a potential therapeutic medicine, appears capable of controlling neuronal hyperexcitability in Mecp2-/Y mice, which occurs in the absence of THIP in the recording solution, lasts at least 1 week after withdrawal, and may contribute to the RTT-like symptom mitigation.


Subject(s)
Action Potentials/drug effects , GABA-A Receptor Agonists/administration & dosage , Isoxazoles/administration & dosage , Locus Coeruleus/drug effects , Neurons/drug effects , Rett Syndrome/physiopathology , Tegmentum Mesencephali/drug effects , Animals , Apnea/physiopathology , Apnea/prevention & control , Disease Models, Animal , Female , GABA-A Receptor Agonists/therapeutic use , Isoxazoles/therapeutic use , Locus Coeruleus/physiopathology , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Receptors, GABA-A , Rett Syndrome/prevention & control , Tegmentum Mesencephali/physiopathology
7.
J Cell Physiol ; 232(5): 1151-1164, 2017 05.
Article in English | MEDLINE | ID: mdl-27670841

ABSTRACT

People with Rett syndrome (RTT) have defects in motor function also seen in Mecp2-null mice. Motor function depends on not only central motor commands but also sensory feedback that is vulnerable to changes in excitability of propriosensory neurons. Here we report evidence for hyperexcitability of mesencephalic trigeminal (Me5) neurons in Mecp2-null mice and a novel cellular mechanism for lowering its impact. In in vitro brain slices, the Me5 neurons in both Mecp2-/Y male and symptomatic Mecp2+/- female mice were overly excitable showing increased firing activity in comparison to their wild-type (WT) male and asymptomatic counterparts. In Mecp2-/Y males, Me5 neurons showed a reduced firing threshold. Consistently, the steady-state activation of voltage-gated Na+ currents (INa ) displayed a hyperpolarizing shift in the Mecp2-null neurons with no change in the INa density. This seems to be due to NaV1.1, SCN1B and SCN4B overexpression and NaV1.2 and SCN3B under-expression. In contrast to the hyperexcitability, the sag potential and postinhibitory rebound (PIR) were reduced in Mecp2-null mice. In voltage-clamp, the IH density was deficient by ∼33%, and the steady-state half-activation had a depolarizing shift of ∼10 mV in the Mecp2-null mice. Quantitative PCR analysis indicated that HCN2 was decreased, HCN1 was upregulated with no change in HCN4 in Mecp2-/Y mice compared to WT. Lastly, blocking IH reduced the firing rate much more in WT than in Mecp2-null neurons. These data suggest that the Mecp2 defect causes an increase in Me5 neuronal excitability likely attributable to alterations in INa , meanwhile IH is reduced likely altering neuronal excitability as well. J. Cell. Physiol. 232: 1151-1164, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Ion Channels/metabolism , Membrane Potentials , Mesencephalon/metabolism , Neurons/metabolism , Rett Syndrome/metabolism , Trigeminal Nerve/cytology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Female , Ion Channel Gating/drug effects , Male , Membrane Potentials/drug effects , Mesencephalon/drug effects , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Protein Subunits/metabolism , Pyrimidines/pharmacology , Rett Syndrome/pathology , Sodium Channels/metabolism
8.
Neuropsychopharmacology ; 41(13): 2977-2986, 2016 12.
Article in English | MEDLINE | ID: mdl-27383008

ABSTRACT

Urges to eat are influenced by stimuli in the environment that are associated with food (food cues). Obese people are more sensitive to food cues, reporting stronger craving and consuming larger portions after food cue exposure. The nucleus accumbens (NAc) mediates cue-triggered motivational responses, and activations in the NAc triggered by food cues are stronger in people who are susceptible to obesity. This has led to the idea that alterations in NAc function similar to those underlying drug addiction may contribute to obesity, particularly in obesity-susceptible individuals. Motivational responses are mediated in part by NAc AMPA receptor (AMPAR) transmission, and recent work shows that cue-triggered motivation is enhanced in obesity-susceptible rats after 'junk-food' diet consumption. Therefore, here we determined whether NAc AMPAR expression and function is increased by 'junk-food' diet consumption in obesity-susceptible vs -resistant populations using both outbred and selectively bred models of susceptibility. In addition, cocaine-induced locomotor activity was used as a general 'read out' of mesolimbic function after 'junk-food' consumption. We found a sensitized locomotor response to cocaine in rats that gained weight on a 'junk-food' diet, consistent with greater responsivity of mesolimbic circuits in obesity-susceptible groups. In addition, eating 'junk-food' increased NAc calcium-permeable-AMPAR (CP-AMPAR) function only in obesity-susceptible rats. This increase occurred rapidly, persisted for weeks after 'junk-food' consumption ceased, and preceded the development of obesity. These data are considered in light of enhanced cue-triggered motivation and striatal function in obesity-susceptible rats and the role of NAc CP-AMPARs in enhanced motivation and addiction.


Subject(s)
Behavior, Addictive/physiopathology , Cues , Food , Motivation/physiology , Nucleus Accumbens/metabolism , Receptors, AMPA/metabolism , Animals , Behavior, Addictive/drug therapy , Behavior, Addictive/psychology , Calcium/metabolism , Cocaine/pharmacology , Disease Models, Animal , Dopamine Uptake Inhibitors/pharmacology , Feeding Behavior , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Locomotion/drug effects , Male , Obesity/pathology , Obesity/psychology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics
9.
PLoS One ; 11(1): e0146470, 2016.
Article in English | MEDLINE | ID: mdl-26785258

ABSTRACT

The locus coeruleus (LC)-norepinephrine (NE) system in the brainstem plays a critical role in a variety of behaviors is an important target of pharmacological intervention to several neurological disorders. Although GABA is the major inhibitory neurotransmitter of LC neurons, the modulation of LC neuronal firing activity by local GABAergic interneurons remains poorly understood with respect to their precise location, intrinsic membrane properties and synaptic modulation. Here, we took an optogenetic approach to address these questions. Channelrhodopsin (ChR2) in a tandem with the yellow fluorescent protein (YFP) was expressed in GABAergic neurons under the control of glutamic acid decarboxylase 2 (GAD2) promoter. Immediately dorsomedial to the LC nucleus, a group of GABAergic neurons was observed. They had small soma and were densely packed in a small area, which we named the dorsomedial LC or dmLC nucleus. These GABAergic neurons showed fast firing activity, strong inward rectification and spike frequency adaptation. Lateral inhibition among these GABAergic neurons was observed. Optostimulation of the dmLC area drastically inhibited LC neuronal firing frequency, expanded the spike intervals, and reset their pacemaking activity. Analysis of the light evoked inhibitory postsynaptic currents (IPSCs) indicated that they were monosynaptic. Such light evoked IPSCs were not seen in slices where this group of GABAergic neurons was absent. Thus, an isolated group of GABAergic neurons is demonstrated in the LC area, whose location, somatic morphology and intrinsic membrane properties are clearly distinguishable from adjacent LC neurons. They interact with each and may inhibit LC neurons as well as a part of local neuronal circuitry in the LC.


Subject(s)
GABAergic Neurons/cytology , Locus Coeruleus/cytology , Animals , Cell Separation , Cell Tracking , Electrophysiological Phenomena , GABAergic Neurons/physiology , Interneurons/cytology , Interneurons/physiology , Locus Coeruleus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics , Patch-Clamp Techniques , Synaptic Transmission/physiology
10.
Psychopharmacology (Berl) ; 233(5): 773-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26612617

ABSTRACT

RATIONALE: Basal and diet-induced differences in mesolimbic function, particularly within the nucleus accumbens (NAc), may contribute to human obesity; these differences may be more pronounced in susceptible populations. OBJECTIVES: We examined differences in cocaine-induced behavioral plasticity in rats that are susceptible vs. resistant to diet-induced obesity and basal differences in striatal neuron function in adult and in adolescent obesity-prone and obesity-resistant rats. METHODS: Susceptible and resistant outbred rats were identified based on "junk-food" diet-induced obesity. Then, the induction and expression of cocaine-induced locomotor sensitization, which is mediated by enhanced striatal function and is associated with increased motivation for rewards and reward-paired cues, were evaluated. Basal differences in mesolimbic function were examined in selectively bred obesity-prone and obesity-resistant rats (P70-80 and P30-40) using both cocaine-induced locomotion and whole-cell patch clamping approaches in NAc core medium spiny neurons (MSNs). RESULTS: In rats that became obese after eating junk-food, the expression of locomotor sensitization was enhanced compared to non-obese rats, with similarly strong responses to 7.5 and 15 mg/kg cocaine. Without diet manipulation, obesity-prone rats were hyper-responsive to the acute locomotor-activating effects of cocaine, and the intrinsic excitability of NAc core MSNs was enhanced by ∼60 % at positive and negative potentials. These differences were present in adult, but not adolescent rats. Post-synaptic glutamatergic transmission was similar between groups. CONCLUSIONS: Mesolimbic systems, particularly NAc MSNs, are hyper-responsive in obesity-prone individuals, and interactions between predisposition and experience influence neurobehavioral plasticity in ways that may promote weight gain and hamper weight loss in susceptible rats.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Nucleus Accumbens/drug effects , Obesity/physiopathology , Aging , Animals , Diet , Dose-Response Relationship, Drug , Glutamates/physiology , Male , Neostriatum/cytology , Neostriatum/drug effects , Nucleus Accumbens/cytology , Obesity/genetics , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
11.
J Physiol Sci ; 65(5): 451-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26071253

ABSTRACT

Rett syndrome (RTT) is a female neurodevelopmental disease with breathing abnormalities. To understand whether breathing defects occur in the early lives of a group of female Mecp2(+/-) mice, a mouse model of RTT, and what percentage of mice shows RTT-like breathing abnormality, breathing activity was measured by plethysmography in conscious mice. Breathing frequency variation and central apnea in a group of Mecp2(+/-) females displayed a distribution pattern similar to Mecp2(-/Y) males, while the rest resembled the wild-type mice. Similar results were obtained using the k-mean clustering statistics analysis. With two independent methods, about 20% of female Mecp2(+/-) mice showed RTT-like breathing abnormalities that began as early as 3 weeks of age in the Mecp2(+/-) mice, and were suppressed with 3% CO2. The finding that only a small proportion of Mecp2(+/-) mice develops RTT-like breathing abnormalities suggests incomplete allele inactivation in the RTT-model Mecp2(+/-) mice.


Subject(s)
Lung/physiopathology , Respiration , Rett Syndrome/physiopathology , Administration, Inhalation , Age Factors , Animals , Apnea/genetics , Apnea/physiopathology , Carbon Dioxide/administration & dosage , Disease Models, Animal , Female , Genetic Predisposition to Disease , Heterozygote , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mice, Knockout , Phenotype , Plethysmography , Respiratory Rate , Rett Syndrome/genetics , Rett Syndrome/therapy
12.
J Biol Chem ; 290(30): 18400-11, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-25979331

ABSTRACT

People with Rett syndrome and mouse models show autonomic dysfunction involving the brain stem locus coeruleus (LC). Neurons in the LC of Mecp2-null mice are overly excited, likely resulting from a defect in neuronal intrinsic membrane properties and a deficiency in GABA synaptic inhibition. In addition to the synaptic GABA receptors, there is a group of GABAA receptors (GABAARs) that is located extrasynaptically and mediates tonic inhibition. Here we show evidence for augmentation of the extrasynaptic GABAARs in Mecp2-null mice. In brain slices, exposure of LC neurons to GABAAR agonists increased tonic currents that were blocked by GABAAR antagonists. With 10 µm GABA, the bicuculline-sensitive tonic currents were ∼4-fold larger in Mecp2-null LC neurons than in the WT. Single-cell PCR analysis showed that the δ subunit, the principal subunit of extrasynaptic GABAARs, was present in LC neurons. Expression levels of the δ subunit were ∼50% higher in Mecp2-null neurons than in the WT. Also increased in expression in Mecp2-null mice was another extrasynaptic GABAAR subunit, α6, by ∼4-fold. The δ subunit-selective agonists 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol hydrochloride and 4-chloro-N-[2-(2-thienyl)imidazo[1,2-a]pyridin-3-yl]]benzamide activated the tonic GABAA currents in LC neurons and reduced neuronal excitability to a greater degree in Mecp2-null mice than in the WT. Consistent with these findings, in vivo application of 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol hydrochloride alleviated breathing abnormalities of conscious Mecp2-null mice. These results suggest that extrasynaptic GABAARs seem to be augmented with Mecp2 disruption, which may be a compensatory response to the deficiency in GABAergic synaptic inhibition and allows control of neuronal excitability and breathing abnormalities.


Subject(s)
GABAergic Neurons/metabolism , Methyl-CpG-Binding Protein 2/genetics , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Rett Syndrome/genetics , Animals , Bicuculline/administration & dosage , GABA Agonists/administration & dosage , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Humans , Isoxazoles/administration & dosage , Locus Coeruleus/metabolism , Locus Coeruleus/physiopathology , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Transgenic , Receptors, GABA-A/genetics , Respiration/genetics , Rett Syndrome/physiopathology , Single-Cell Analysis , Synapses/drug effects , Synapses/metabolism
13.
Am J Physiol Cell Physiol ; 307(6): C508-20, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25009110

ABSTRACT

Rett syndrome is an autism-spectrum disorder resulting from mutations to the X-linked gene, methyl-CpG binding protein 2 (MeCP2), which causes abnormalities in many systems. It is possible that the body may develop certain compensatory mechanisms to alleviate the abnormalities. The norepinephrine system originating mainly in the locus coeruleus (LC) is defective in Rett syndrome and Mecp2-null mice. LC neurons are subject to modulation by GABA, glutamate, and acetylcholine (ACh), providing an ideal system to test the compensatory hypothesis. Here we show evidence for potential compensatory modulation of LC neurons by post- and presynaptic ACh inputs. We found that the postsynaptic currents of nicotinic ACh receptors (nAChR) were smaller in amplitude and longer in decay time in the Mecp2-null mice than in the wild type. Single-cell PCR analysis showed a decrease in the expression of α3-, α4-, α7-, and ß3-subunits and an increase in the α5- and α6-subunits in the mutant mice. The α5-subunit was present in many of the LC neurons with slow-decay nAChR currents. The nicotinic modulation of spontaneous GABAA-ergic inhibitory postsynaptic currents in LC neurons was enhanced in Mecp2-null mice. In contrast, the nAChR manipulation of glutamatergic input to LC neurons was unaffected in both groups of mice. Our current-clamp studies showed that the modulation of LC neurons by ACh input was reduced moderately in Mecp2-null mice, despite the major decrease in nAChR currents, suggesting possible compensatory processes may take place, thus reducing the defects to a lesser extent in LC neurons.


Subject(s)
Acetylcholine/metabolism , Cholinergic Neurons/metabolism , Locus Coeruleus/metabolism , Rett Syndrome/metabolism , Adaptation, Physiological , Animals , Disease Models, Animal , Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials , Kinetics , Locus Coeruleus/physiopathology , Male , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Receptors, GABA-A/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Receptors, Presynaptic/genetics , Receptors, Presynaptic/metabolism , Rett Syndrome/genetics , Rett Syndrome/physiopathology , gamma-Aminobutyric Acid/metabolism
14.
J Comp Neurol ; 518(3): 255-76, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19941347

ABSTRACT

Dopamine (DA) modulates motor systems in phyla as diverse as nematodes and arthropods up through chordates. A comparison of dopaminergic systems across a broad phylogenetic range should reveal shared organizing principles. The pyloric network, located in the stomatogastric ganglion (STG), is an important model for neuromodulation of motor networks. The effects of DA on this network have been well characterized at the circuit and cellular levels in the spiny lobster, Panulirus interruptus. Here we provide the first data about the physical organization of the DA signaling system in the STG and the function of D(2) receptors in pyloric neurons. Previous studies showed that DA altered intrinsic firing properties and synaptic output in the pyloric dilator (PD) neuron, in part by reducing calcium currents and increasing outward potassium currents. We performed single cell reverse transcriptase-polymerase chain reaction (RT-PCR) experiments to show that PD neurons exclusively expressed a type 2 (D(2alphaPan)) DA receptor. This was confirmed by using confocal microscopy in conjunction with immunohistochemistry (IHC) on STG whole-mount preparations containing dye-filled PD neurons. Immunogold electron microscopy showed that surface receptors were concentrated in fine neurites/terminal swellings and vesicle-laden varicosities in the synaptic neuropil. Double-label IHC experiments with tyrosine hydroxylase antiserum suggested that the D(2alphaPan) receptors received volume neurotransmissions. Receptors were further mapped onto three-dimensional models of PD neurons built from Neurolucida tracings of confocal stacks from the IHC experiments. The data showed that D(2alphaPan) receptors were selectively targeted to approximately 40% of synaptic structures in any given PD neuron, and were nonuniformly distributed among neurites.


Subject(s)
Enteric Nervous System/metabolism , Ganglia, Invertebrate/metabolism , Neurons/metabolism , Palinuridae/metabolism , Receptors, Dopamine D2/genetics , Synapses/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Cell Shape/physiology , Dopamine/biosynthesis , Enteric Nervous System/ultrastructure , Fluorescent Dyes , Ganglia, Invertebrate/ultrastructure , Gastrointestinal Tract/innervation , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Confocal , Microscopy, Immunoelectron , Neurons/ultrastructure , Palinuridae/ultrastructure , Paracrine Communication/physiology , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Synapses/ultrastructure , Synaptic Transmission/physiology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...