Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
CNS Neurosci Ther ; 30(1): e14468, 2024 01.
Article in English | MEDLINE | ID: mdl-37950551

ABSTRACT

AIMS: This study aimed to investigate the effect of perineuronal net (PNN) and neurocan (NCAN) on spinal inhibitory parvalbumin interneuron (PV-IN), and the mechanism of electroacupuncture (EA) in promoting spinal cord injury (SCI) repair through neurocan in PNN. METHODS: A mouse model of SCI was established. Sham-operated mice or SCI model mice were treated with chondroitin sulfate ABC (ChABC) enzyme or control vehicle for 2 weeks (i.e., sham+veh group, sham+ChABC group, SCI+veh group, and SCI+ChABC group, respectively), and then spinal cord tissues were taken from the T10 lesion epicenter for RNA sequencing (RNA-seq). MSigDB Hallmark and C5 databases for functional analysis, analysis strategies such as differential expression gene analysis (DEG), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI). According to the results of RNA-seq analysis, the expression of NCAN was knocked down or overexpressed by virus intervention, or/and EA intervention. Polymerase chain reaction (PCR), immunofluorescence, western blot, electrophysiological, and behavioral tests were performed. RESULTS: After the successful establishment of SCI model, the motor dysfunction of lower limbs, and the expression of PNN core glycan protein at the epicenter of SCI were reduced. RNA-seq and PCR showed that PNN core proteoglycans except NCAN showed the same expression trend in normal and injured spinal cord treated with ChABC. KEGG and GSEA showed that PNN is mainly associated with inhibitory GABA neuronal function in injured spinal cord tissue, and PPI showed that NCAN in PNN can be associated with inhibitory neuronal function through parvalbumin (PV). Calcium imaging showed that local parvalbumin interneuron (PV-IN) activity decreased after PNN destruction, whether due to ChABC treatment or surgical bruising of the spinal cord. Overexpression of neurocan in injured spinal cord can enhance local PV-IN activity. PCR and western blot suggested that overexpression or knockdown of neurocan could up-regulate or down-regulate the expression of GAD. At the same time, the activity of PV-IN in the primary motor cortex (M1) and the primary sensory cortex of lower (S1HL) extremity changed synchronously. In addition, overexpression of neurocan improved the electrical activity of the lower limb and promoted functional repair of the paralyzed hind limb. EA intervention reversed the down-regulation of neurocan, enhanced the expression of PNN in the lesioned area, M1 and S1HL. CONCLUSION: Neurocan in PNN can regulate the activity of PV-IN, and EA can promote functional recovery of mice with SCI by upregulating neurocan expression in PNN.


Subject(s)
Electroacupuncture , Spinal Cord Injuries , Animals , Mice , Rats , GABAergic Neurons/metabolism , Neurocan , Parvalbumins/metabolism , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord Injuries/pathology
2.
Addict Biol ; 28(9): e13314, 2023 09.
Article in English | MEDLINE | ID: mdl-37644891

ABSTRACT

Methamphetamine (Meth) withdrawal elicits anxiety, which is a public health concern with limited therapeutic options. Previous studies implied a strong correlation between mPFC and Meth withdrawal. Here, we examined the role of Gegen-Qinlian decoction (GQD) in Meth withdrawal anxiety and explored potential therapeutic targets in mPFC. We found that intra-gastric administration of GQD during the withdrawal period efficiently alleviated anxiety-like behaviours in Meth-withdrawn mice. Further, GQD could restore Meth withdrawal-triggered pathway of GABAergic interneurons (GABA IN)-pyramidal neurons (PN) in the mPFC of Meth-withdrawn mice, especially the prelimbic cortex (PrL) sub-region and PV-positive GABA IN. While, GQD had no obvious effects on the glial cells in the mPFC of Meth-withdrawn mice. By transcriptomic analysis and validation of several gene candidates, we found that genes in the MAPK signalling pathway, especially those related to heat shock proteins, including Hspa1a, Hspa1b and Hspb1, might be GQD-targeting genes in mPFC to treat Meth withdrawal anxiety, as indicated that these genes were up-regulated by Meth withdrawal but rescued by GQD in mPFC. Collectively, our findings identified for the first time that GQD could efficiently alleviate Meth withdrawal anxiety, partially through regulating the local GABA IN-PN pathway and transcriptomic profile of mPFC. The present study confirms that TCM, such as GQD, will be a desirable therapeutic approach in the treatment of drug addiction and related emotional deficits.


Subject(s)
Amphetamine-Related Disorders , Methamphetamine , Substance Withdrawal Syndrome , Animals , Mice , Medicine, Chinese Traditional , Anxiety/drug therapy , Pyramidal Cells , Substance Withdrawal Syndrome/drug therapy , Interneurons , gamma-Aminobutyric Acid
3.
Elife ; 112022 Nov 07.
Article in English | MEDLINE | ID: mdl-36342840

ABSTRACT

Axons of retinal ganglion cells (RGCs) play critical roles in the development of inhibitory circuits in visual thalamus. We previously reported that RGC axons signal astrocytes to induce the expression of fibroblast growth factor 15 (FGF15), a motogen required for GABAergic interneuron migration into visual thalamus. However, how retinal axons induce thalamic astrocytes to generate Fgf15 and influence interneuron migration remains unknown. Here, we demonstrate that impairing RGC activity had little impact on interneuron recruitment into mouse visual thalamus. Instead, our data show that retinal-derived sonic hedgehog (SHH) is essential for interneuron recruitment. Specifically, we show that thalamus-projecting RGCs express SHH and thalamic astrocytes generate downstream components of SHH signaling. Deletion of RGC-derived SHH leads to a significant decrease in Fgf15 expression, as well as in the percentage of interneurons recruited into visual thalamus. Overall, our findings identify a morphogen-dependent neuron-astrocyte signaling mechanism essential for the migration of thalamic interneurons.


Subject(s)
Hedgehog Proteins , Interneurons , Mice , Animals , Hedgehog Proteins/metabolism , Interneurons/physiology , Thalamus/metabolism , Axons/metabolism , Retinal Ganglion Cells/metabolism
4.
Brain Res Bull ; 187: 181-198, 2022 09.
Article in English | MEDLINE | ID: mdl-35850189

ABSTRACT

Sleep abnormalities are widely reported in patients with Alzheimer's disease (AD) and are linked to cognitive impairments. Sleep abnormalities could be potential biomarkers to detect AD since they are often observed at the preclinical stage. Moreover, sleep could be a target for early intervention to prevent or slow AD progression. Thus, here we review changes in brain oscillations observed during sleep, their connection to AD pathophysiology and the role of specific brain circuits. Slow oscillations (0.1-1 Hz), sleep spindles (8-15 Hz) and their coupling during non-REM sleep are consistently reduced in studies of patients and in AD mouse models although the timing and magnitude of these alterations depends on the pathophysiological changes and the animal model studied. Changes in delta (1-4 Hz) activity are more variable. Animal studies suggest that hippocampal sharp-wave ripples (100-250 Hz) are also affected. Reductions in REM sleep amount and slower oscillations during REM are seen in patients but less consistently in animal models. Thus, changes in a variety of sleep oscillations could impact sleep-dependent memory consolidation or restorative functions of sleep. Recent mechanistic studies suggest that alterations in the activity of GABAergic neurons in the cortex, hippocampus and thalamic reticular nucleus mediate sleep oscillatory changes in AD and represent a potential target for intervention. Longitudinal studies of the timing of AD-related sleep abnormalities with respect to pathology and dysfunction of specific neural networks are needed to identify translationally relevant biomarkers and guide early intervention strategies to prevent or delay AD progression.


Subject(s)
Alzheimer Disease , GABAergic Neurons , Animals , Electroencephalography , GABAergic Neurons/physiology , Hippocampus/physiology , Mice , Sleep/physiology , Thalamus/physiology
5.
Cereb Cortex ; 32(13): 2785-2796, 2022 06 16.
Article in English | MEDLINE | ID: mdl-34689202

ABSTRACT

Gamma oscillations have received considerable attention owing to their association with cognitive function and various neuropsychiatric disorders. However, interactions of gamma oscillations at different frequency bands in humans remain unclear. In the present magnetoencephalographic study, brain oscillations in a wide frequency range were examined using a time-frequency analysis during the 20-, 30-, 40-, and 50-Hz auditory stimuli in 21 healthy subjects. First, dipoles for auditory steady-state response (ASSR) were estimated and interaction among oscillations at 10-60 Hz was examined using the source strength waveforms. Results showed the suppression of ongoing low-gamma oscillations at approximately 30 Hz during stimulation at 40 Hz. Second, multi-dipole analyses suggested that the main dipole for ASSR and dipoles for suppressed low-frequency gamma oscillations were distinct. Third, an all-sensor analysis was performed to clarify the distribution of the 40-Hz ASSR and suppression of low-frequency gamma oscillations. Notably, the area of suppression surrounded the center of the 40-Hz ASSR and showed a trend of extending to the vertex, indicating that different groups of neurons were responsible for these two gamma oscillations and that the 40-Hz oscillation circuit have specific inhibitory innervation to the low-gamma circuit.


Subject(s)
Auditory Cortex , Evoked Potentials, Auditory , Acoustic Stimulation/methods , Auditory Cortex/physiology , Electroencephalography/methods , Evoked Potentials, Auditory/physiology , Gamma Rhythm/physiology , Humans , Magnetoencephalography/methods , Physical Therapy Modalities
6.
Cell Rep ; 37(3): 109837, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686328

ABSTRACT

The selection of goal-directed behaviors is supported by neural circuits located within the frontal cortex. Frontal cortical afferents arise from multiple brain areas, yet the cell-type-specific targeting of these inputs is unclear. Here, we use monosynaptic retrograde rabies mapping to examine the distribution of afferent neurons targeting distinct classes of local inhibitory interneurons and excitatory projection neurons in mouse infralimbic frontal cortex. Interneurons expressing parvalbumin, somatostatin, or vasoactive intestinal peptide receive a large proportion of inputs from the hippocampus, while interneurons expressing neuron-derived neurotrophic factor receive a large proportion of inputs from thalamic regions. A similar dichotomy is present among the four different excitatory projection neurons. These results show a prominent bias among long-range hippocampal and thalamic afferent systems in their targeting to specific sets of frontal cortical neurons. Moreover, they suggest the presence of two distinct local microcircuits that control how different inputs govern frontal cortical information processing.


Subject(s)
Frontal Lobe/physiology , Hippocampus/physiology , Interneurons/physiology , Synapses/physiology , Thalamus/physiology , Animals , Behavior, Animal , Frontal Lobe/cytology , Frontal Lobe/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neural Inhibition , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Parvalbumins/genetics , Parvalbumins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Synapses/metabolism , Thalamus/cytology , Thalamus/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
7.
Neuron ; 109(17): 2682-2690.e5, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34314698

ABSTRACT

Slow-wave sleep is characterized by near-synchronous alternation of active Up states and quiescent Down states in the neocortex. Although the cortex itself can maintain these oscillations, the full expression of Up-Down states requires intact thalamocortical circuits. Sensory thalamic input can drive the cortex into an Up state. Here we show that midline thalamic neurons terminate Up states synchronously across cortical areas. Combining local field potential, single-unit, and patch-clamp recordings in conjunction with optogenetic stimulation and silencing in mice in vivo, we report that thalamic input mediates Down transition via activation of layer 1 neurogliaform inhibitory neurons acting on GABAB receptors. These results strengthen the evidence that thalamocortical interactions are essential for the full expression of slow-wave sleep, show that Down transition is an active process mediated by cortical GABAB receptors, and demonstrate that thalamus synchronizes Down transitions across cortical areas during natural slow-wave sleep.


Subject(s)
Interneurons/physiology , Neocortex/physiology , Receptors, GABA-B/metabolism , Sleep, Slow-Wave/physiology , Thalamus/physiology , Animals , Evoked Potentials , Female , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/metabolism , Thalamus/cytology , Thalamus/metabolism
8.
Elife ; 102021 02 04.
Article in English | MEDLINE | ID: mdl-33538694

ABSTRACT

Three rules govern the connectivity between neurons in the thalamus and inhibitory neurons in the visual cortex of rabbits.


Subject(s)
Visual Cortex , Animals , Neurons , Rabbits , Thalamus
9.
Mol Brain ; 14(1): 33, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33588899

ABSTRACT

GABAergic interneurons play a critical role in tuning neural networks in the central nervous system, and their defects are associated with neuropsychiatric disorders. Currently, the mDlx enhancer is solely used for adeno-associated virus (AAV) vector-mediated transgene delivery into cortical interneurons. Here, we developed a new inhibitory neuron-specific promoter (designated as the mGAD65 promoter), with a length of 2.5 kb, from a mouse genome upstream of exon 1 of the Gad2 gene encoding glutamic acid decarboxylase (GAD) 65. Intravenous infusion of blood-brain barrier-penetrating AAV-PHP.B expressing an enhanced green fluorescent protein under the control of the mGAD65 promoter transduced the whole brain in an inhibitory neuron-specific manner. The specificity and efficiency of the mGAD65 promoter for GABAergic interneurons, which was assessed at the motor cortex, were almost identical to or slightly higher than those of the mDlx enhancer. Immunohistochemical analysis revealed that the mGAD65 promoter preferentially transduced parvalbumin (PV)-expressing interneurons. Notably, the mGAD65 promoter transduced chandelier cells more efficiently than the mDlx enhancer and robustly labeled their synaptic boutons, called the cartridge, targeting the axon initial segments of excitatory pyramidal neurons. To test the ability of the mGAD65 promoter to express a functional molecule, we virally expressed G-CaMP, a fluorescent Ca2+ indicator, in the motor cortex, and this enabled us to monitor spontaneous and drug-induced Ca2+ activity in GABAergic inhibitory neurons. These results suggest that the mGAD65 promoter is useful for AAV-mediated targeting and manipulation of GABAergic neurons with the dominance of cortical PV-expressing neurons, including chandelier cells.


Subject(s)
Brain/metabolism , Dependovirus/metabolism , GABAergic Neurons/metabolism , Plasmids/metabolism , Transduction, Genetic , Animals , Calcium/metabolism , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Injections, Intravenous , Interneurons/metabolism , Mice, Inbred C57BL , Motor Cortex/metabolism , Neurons/metabolism , Parvalbumins/metabolism , Promoter Regions, Genetic
10.
Acta Neuropathol Commun ; 8(1): 208, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33256853

ABSTRACT

Alcohol affects multiple neurotransmitter systems, notably the GABAergic system and has been recognised for a long time as particularly damaging during critical stages of brain development. Nevertheless, data from the literature are most often derived from animal or in vitro models. In order to study the production, migration and cortical density disturbances of GABAergic interneurons upon prenatal alcohol exposure, we performed immunohistochemical studies by means of the proliferation marker Ki67, GABA and calretinin antibodies in the frontal cortical plate of 17 foetal and infant brains antenatally exposed to alcohol, aged 15 weeks' gestation to 22 postnatal months and in the ganglionic eminences and the subventricular zone of the dorsal telencephalon until their regression, i.e., 34 weeks' gestation. Results were compared with those obtained in 17 control brains aged 14 weeks of gestation to 35 postnatal months. We also focused on interneuron vascular migration along the cortical microvessels by confocal microscopy with double immunolabellings using Glut1, GABA and calretinin. Semi-quantitative and quantitative analyses of GABAergic and calretininergic interneuron density allowed us to identify an insufficient and delayed production of GABAergic interneurons in the ganglionic eminences during the two first trimesters of the pregnancy and a delayed incorporation into the laminar structures of the frontal cortex. Moreover, a mispositioning of GABAergic and calretininergic interneurons persisted throughout the foetal life, these cells being located in the deep layers instead of the superficial layers II and III. Moreover, vascular migration of calretininergic interneurons within the cortical plate was impaired, as reflected by low numbers of interneurons observed close to the cortical perforating vessel walls that may in part explain their abnormal intracortical distribution. Our results are globally concordant with those previously obtained in mouse models, in which alcohol has been shown to induce an interneuronopathy by affecting interneuron density and positioning within the cortical plate, and which could account for the neurological disabilities observed in children with foetal alcohol disorder spectrum.


Subject(s)
Alcohol Drinking , Brain/embryology , Calbindin 2/metabolism , Fetal Alcohol Spectrum Disorders/metabolism , Fetus/embryology , Interneurons/metabolism , Ki-67 Antigen/metabolism , Prenatal Exposure Delayed Effects/metabolism , gamma-Aminobutyric Acid/metabolism , Alcoholism , Binge Drinking , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Movement , Female , Fetal Alcohol Spectrum Disorders/pathology , Fetus/metabolism , Fetus/pathology , Frontal Lobe/embryology , Frontal Lobe/metabolism , Frontal Lobe/pathology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Humans , Infant , Infant, Newborn , Interneurons/pathology , Male , Pregnancy , Pregnancy Complications , Pregnancy Trimester, Second , Prenatal Exposure Delayed Effects/pathology , Telencephalon/embryology , Telencephalon/metabolism , Telencephalon/pathology
11.
FASEB J ; 34(9): 11913-11924, 2020 09.
Article in English | MEDLINE | ID: mdl-32683743

ABSTRACT

We recently found that adolescent cocaine exposure (ACE) resulted in an enhancement of the γ-aminobutyric acid (GABA) neurotransmitter system in the prelimbic cortex (PrL) of adult mice. Here, we aim to further investigate the role of GABAergic transmission, especially parvalbumin (PV) interneurons within PrL in the development of ACE-induced anxiety-like behavior, and to assess whether and how electro-acupuncture (EA) therapeutically manage the ACE-induced abnormal behaviors in adulthood. ACE mice exhibited the enhanced anxiety-like behaviors in their adulthood, accompanied by increased GABAergic transmission and PV interneurons in PrL. Chemogenetic blocking PV interneurons in PrL alleviated ACE-enhanced anxiety-like behaviors in mice. Importantly, 37-day EA treatments (mixture of 2 Hz/100 Hz, 1 mA, 30 minutes once a day) at the acupoints of Yintang (GV29) and Baihui (GV20) also alleviated ACE-induced anxiety-like behaviors, and rescued ACE-impaired GABAergic neurotransmitter system and PV interneurons in PrL. In parallel, EA treatments further suppressed the activities of pyramidal neurons in PrL, suggesting that EA treatments seem to perform it beneficial effects on the ACE-induced abnormal emotional behaviors by "calming down" the whole PrL. Collectively, these findings revealed that hyper-function of GABAergic transmission, especially mediating by PV interneurons in PrL may be key etiology underlying ACE-induced anxiety-like behaviors. At least by normalizing the function of GABAergic and PV interneurons, EA may represent a promising therapeutic strategy for managing adolescent substance use-related emotional disorders.


Subject(s)
Anxiety , Behavior, Animal , Cocaine-Related Disorders , Electroacupuncture , Interneurons/metabolism , Parvalbumins/metabolism , Animals , Anxiety/metabolism , Anxiety/physiopathology , Anxiety/therapy , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/therapy , Limbic System/metabolism , Limbic System/physiopathology , Male , Mice , Mice, Transgenic
12.
Front Neural Circuits ; 14: 13, 2020.
Article in English | MEDLINE | ID: mdl-32296311

ABSTRACT

In the neocortex, neuronal processing of sensory events is significantly influenced by context. For instance, responses in sensory cortices are suppressed to repetitive or redundant stimuli, a phenomenon termed "stimulus-specific adaptation" (SSA). However, in a context in which that same stimulus is novel, or deviates from expectations, neuronal responses are augmented. This augmentation is termed "deviance detection" (DD). This contextual modulation of neural responses is fundamental for how the brain efficiently processes the sensory world to guide immediate and future behaviors. Notably, context modulation is deficient in some neuropsychiatric disorders such as schizophrenia (SZ), as quantified by reduced "mismatch negativity" (MMN), an electroencephalography waveform reflecting a combination of SSA and DD in sensory cortex. Although the role of NMDA-receptor function and other neuromodulatory systems on MMN is established, the precise microcircuit mechanisms of MMN and its underlying components, SSA and DD, remain unknown. When coupled with animal models, the development of powerful precision neurotechnologies over the past decade carries significant promise for making new progress into understanding the neurobiology of MMN with previously unreachable spatial resolution. Currently, rodent models represent the best tool for mechanistic study due to the vast genetic tools available. While quantifying human-like MMN waveforms in rodents is not straightforward, the "oddball" paradigms used to study it in humans and its underlying subcomponents (SSA/DD) are highly translatable across species. Here we summarize efforts published so far, with a focus on cortically measured SSA and DD in animals to maintain relevance to the classically measured MMN, which has cortical origins. While mechanistic studies that measure and contrast both components are sparse, we synthesize a potential set of microcircuit mechanisms from the existing rodent, primate, and human literature. While MMN and its subcomponents likely reflect several mechanisms across multiple brain regions, understanding fundamental microcircuit mechanisms is an important step to understand MMN as a whole. We hypothesize that SSA reflects adaptations occurring at synapses along the sensory-thalamocortical pathways, while DD depends on both SSA inherited from afferent inputs and resulting disinhibition of non-adapted neurons arising from the distinct physiology and wiring properties of local interneuronal subpopulations and NMDA-receptor function.


Subject(s)
Acoustic Stimulation/methods , Auditory Cortex/physiology , Evoked Potentials, Auditory/physiology , Nerve Net/physiology , Synapses/physiology , Acoustic Stimulation/psychology , Animals , Electroencephalography/methods , Electroencephalography/psychology , Humans , Receptors, N-Methyl-D-Aspartate/physiology
13.
Neuron ; 106(3): 468-481.e2, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32142646

ABSTRACT

One way to assess a neuron's function is to describe all its inputs and outputs. With this goal in mind, we used serial section electron microscopy to map 899 synaptic inputs and 623 outputs in one inhibitory interneuron in a large volume of the mouse visual thalamus. This neuron innervated 256 thalamocortical cells spread across functionally distinct subregions of the visual thalamus. All but one of its neurites were bifunctional, innervating thalamocortical and local interneurons while also receiving synapses from the retina. We observed a wide variety of local synaptic motifs. While this neuron innervated many cells weakly, with single en passant synapses, it also deployed specialized branches that climbed along other dendrites to form strong multi-synaptic connections with a subset of partners. This neuron's diverse range of synaptic relationships allows it to participate in a mix of global and local processing but defies assigning it a single circuit function.


Subject(s)
Interneurons/physiology , Neural Inhibition , Synapses/physiology , Thalamus/cytology , Visual Cortex/cytology , Animals , Interneurons/cytology , Mice , Mice, Inbred C57BL , Models, Neurological , Neuroanatomical Tract-Tracing Techniques , Thalamus/physiology , Visual Cortex/physiology
14.
Schizophr Res ; 218: 93-98, 2020 04.
Article in English | MEDLINE | ID: mdl-32061454

ABSTRACT

INTRODUCTION: Inhibitory sensory gating of the P50 cerebral evoked potential to paired auditory stimuli (S1, S2) is a widely used paradigm for the study of schizophrenia and related conditions. Its use to measure genetic, treatment, and developmental effects requires a metric with more stable properties than the simple ratio of the paired responses. METHODS: This study assessed the ratio P50S2µV/P50S1µV and P50S2µV co-varied for P50S1µV in all 27 independent published studies that compared schizophrenia patients with healthy controls from 2000 to 2019. The largest study from each research group was selected. The Colorado research group's studies were excluded to eliminate bias from the first report of the phenomenon. RESULTS: Across the 27 studies encompassing 1179 schizophrenia patients and 1091 healthy controls, both P50S2µV co-varied for P50S1µV and P50S2µV/P50S1µV significantly separated the patients from the controls (both P < 0.0001). Effect size for P50S2µV co-varied for P50S1µV is d' = 1.23. The normal distribution of P50S2µV co-varied for P50S1µV detected influences of maternal inflammation and effects on behavior in a recent developmental study, an emerging use for the P50 inhibitory gating measure. P50S2µV/P50S1µV was not normally distributed. Results from two multi-site NIMH genetics collaborations also support the use of P50S2µV as a biomarker. CONCLUSION: Both methods detect an abnormality of cerebral inhibition in schizophrenia with high significance across multiple independent laboratories. The normal distribution of P50S2µV co-varied for P50S1µV makes it more suitable for studies of genetic, treatment, and other influences on the development and expression of inhibitory deficits in schizophrenia.


Subject(s)
Schizophrenia , Acoustic Stimulation , Electroencephalography , Evoked Potentials , Evoked Potentials, Auditory , Humans , Reaction Time , Schizophrenia/genetics , Sensory Gating
15.
Neurosci Bull ; 36(5): 493-505, 2020 May.
Article in English | MEDLINE | ID: mdl-31956963

ABSTRACT

The caudal forelimb area (CFA) of the mouse cortex is essential in many forelimb movements, and diverse types of GABAergic interneuron in the CFA are distinct in the mediation of cortical inhibition in motor information processing. However, their long-range inputs remain unclear. In the present study, we combined the monosynaptic rabies virus system with Cre driver mouse lines to generate a whole-brain map of the inputs to three major inhibitory interneuron types in the CFA. We discovered that each type was innervated by the same upstream areas, but there were quantitative differences in the inputs from the cortex, thalamus, and pallidum. Comparing the locations of the interneurons in two sub-regions of the CFA, we discovered that their long-range inputs were remarkably different in distribution and proportion. This whole-brain mapping indicates the existence of parallel pathway organization in the forelimb subnetwork and provides insight into the inhibitory processes in forelimb movement to reveal the structural architecture underlying the functions of the CFA.


Subject(s)
Brain/anatomy & histology , Forelimb/innervation , GABAergic Neurons , Motor Cortex/anatomy & histology , Animals , Brain Mapping , Cerebellar Cortex/anatomy & histology , Interneurons/physiology , Male , Mice , Neural Pathways/anatomy & histology , Thalamic Diseases/congenital , Thalamus/anatomy & histology
16.
Proc Natl Acad Sci U S A ; 117(5): 2671-2682, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31964831

ABSTRACT

Inhibitory interneurons comprise a fraction of the total neurons in the visual thalamus but are essential for sharpening receptive field properties and improving contrast-gain of retinogeniculate transmission. During early development, these interneurons undergo long-range migration from germinal zones, a process regulated by the innervation of the visual thalamus by retinal ganglion cells. Here, using transcriptomic approaches, we identified a motogenic cue, fibroblast growth factor 15 (FGF15), whose expression in the visual thalamus is regulated by retinal input. Targeted deletion of functional FGF15 in mice led to a reduction in thalamic GABAergic interneurons similar to that observed in the absence of retinal input. This loss may be attributed, at least in part, to misrouting of interneurons into nonvisual thalamic nuclei. Unexpectedly, expression analysis revealed that FGF15 is generated by thalamic astrocytes and not retino-recipient neurons. Thus, these data show that retinal inputs signal through astrocytes to direct the long-range recruitment of interneurons into the visual thalamus.


Subject(s)
Astrocytes/metabolism , Fibroblast Growth Factors/metabolism , Interneurons/metabolism , Thalamus/metabolism , Animals , Fibroblast Growth Factors/genetics , GABAergic Neurons/metabolism , Humans , Mice , Mice, Inbred C57BL , Retina/metabolism , Visual Perception
17.
Cell Rep ; 30(4): 1178-1194.e3, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995757

ABSTRACT

To understand striatal function, it is essential to know the functional organization of the numerous inputs targeting the diverse population of striatal neurons. Using optogenetics, we activated terminals from ipsi- or contralateral primary somatosensory cortex (S1) or primary motor cortex (M1), or thalamus while obtaining simultaneous whole-cell recordings from pairs or triplets of striatal medium spiny neurons (MSNs) and adjacent interneurons. Ipsilateral corticostriatal projections provided stronger excitation to fast-spiking interneurons (FSIs) than to MSNs and only sparse and weak excitation to low threshold-spiking interneurons (LTSIs) and cholinergic interneurons (ChINs). Projections from contralateral M1 evoked the strongest responses in LTSIs but none in ChINs, whereas thalamus provided the strongest excitation to ChINs but none to LTSIs. In addition, inputs varied in their glutamate receptor composition and their short-term plasticity. Our data revealed a highly selective organization of excitatory striatal afferents, which is determined by both pre- and postsynaptic neuronal identity.


Subject(s)
Cholinergic Neurons/physiology , Corpus Striatum/metabolism , Interneurons/physiology , Motor Cortex/physiology , Thalamus/physiology , Animals , Cholinergic Neurons/metabolism , Corpus Striatum/cytology , Corpus Striatum/physiology , Female , Interneurons/metabolism , Male , Mice , Mice, Transgenic , Motor Cortex/radiation effects , Neural Pathways/physiology , Neuronal Plasticity/physiology , Optogenetics , Patch-Clamp Techniques , Receptors, Glutamate/metabolism , Somatosensory Cortex/physiology , Synapses/physiology , Thalamus/radiation effects
18.
Brain Res ; 1727: 146571, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31786200

ABSTRACT

NMDA receptor (NMDAR) antagonists such as ketamine, can reproduce many of the symptoms of schizophrenia. A reliable indicator of NMDAR channel blocker action in vivo is the augmentation of neuronal oscillation power. Since the coordinated and rhythmic activation of neuronal assemblies (oscillations) is necessary for perception, cognition and working memory, their disruption (inappropriate augmentation or inhibition of oscillatory power or inter-regional coherence) both in psychiatric conditions and with NMDAR antagonists may reflect the underlying defects causing schizophrenia symptoms. NMDAR antagonists and knockout (KO) mice were used to evaluate the role of GluN2C and GluN2D NMDAR subunits in generating NMDAR antagonist-induced oscillations. We find that basal oscillatory power was elevated in GluN2C-KO mice, especially in the low gamma frequencies while there was no statistically significant difference in basal oscillations between WT and GluN2D-KO mice. Compared to wildtype (WT) mice, NMDAR channel blockers caused a greater increase in oscillatory power in GluN2C-KO mice and were relatively ineffective in inducing oscillations in GluN2D-KO mice. In contrast, preferential blockade of GluN2A- and GluN2B-containing receptors induced oscillations that did not appear to be changed in either KO animal. We propose a model wherein NMDARs containing GluN2C in astrocytes and GluN2D in interneurons serve to detect local cortical excitatory synaptic activity and provide excitatory and inhibitory feedback, respectively, to local populations of postsynaptic excitatory neurons and thereby bidirectionally modulate oscillatory power.


Subject(s)
Neurofeedback/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Mice , Mice, Knockout , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics
19.
Neuron ; 101(3): 444-458.e6, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30658860

ABSTRACT

The motor symptoms of Parkinson's disease (PD) are thought to stem from an imbalance in the activity of striatal direct- and indirect-pathway spiny projection neurons (SPNs). Disease-induced alterations in the activity of networks controlling SPNs could contribute to this imbalance. One of these networks is anchored by the parafascicular nucleus (PFn) of the thalamus. To determine the role of the PFn in striatal PD pathophysiology, optogenetic, chemogenetic, and electrophysiological tools were used in ex vivo slices from transgenic mice with region-specific Cre recombinase expression. These studies revealed that in parkinsonian mice, the functional connectivity of PFn neurons with indirect pathway SPNs (iSPNs) was selectively enhanced by cholinergic interneurons acting through presynaptic nicotinic acetylcholine receptors (nAChRs) on PFn terminals. Attenuating this network adaptation by chemogenetic or genetic strategies alleviated motor-learning deficits in parkinsonian mice, pointing to a potential new therapeutic strategy for PD patients.


Subject(s)
Cholinergic Neurons/physiology , Corpus Striatum/physiopathology , Excitatory Postsynaptic Potentials , Interneurons/physiology , Parkinson Disease/physiopathology , Thalamus/physiopathology , Animals , Cholinergic Neurons/metabolism , Corpus Striatum/cytology , Glutamic Acid/metabolism , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Parkinson Disease/metabolism , Receptors, Nicotinic/metabolism , Thalamus/cytology
20.
Cereb Cortex ; 29(10): 4107-4118, 2019 09 13.
Article in English | MEDLINE | ID: mdl-30535288

ABSTRACT

GABAergic interneurons perform distinct functions during cortical development in the mouse brain. Among the diverse GABAergic neurons present in the brain, early-born somatostatin (SST)-expressing inhibitory interneurons, which are innervated by other interneurons and local pyramidal cells (PCs), act in a neural computational role in circuitry regulation. The synapses between the SST+ interneurons and other cells form gradually during development. Here, we traced the developmental course of the electrophysiological properties of SST+ interneurons at layer 2/3 of the neocortical secondary motor area (M2) in mouse, and the synaptic connectivity between SST+ interneurons and PCs. Also, we used toxin-mediated and genetic method to suppress the activities of PCs, and demonstrate that decreasing excitatory input at early stage (before P1) rather than late stage (after P8) would delay the functional maturation of SST+ interneurons. In conclusion, our results indicate that early functional activity of PCs is crucial for the intrinsic maturation of SST+ interneurons, following which these interneurons participate in local circuitry.


Subject(s)
Excitatory Postsynaptic Potentials , Interneurons/physiology , Motor Cortex/growth & development , Pyramidal Cells/physiology , Somatostatin/metabolism , Animals , Interneurons/cytology , Interneurons/metabolism , Mice , Motor Cortex/cytology , Motor Cortex/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL