Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.130
1.
Sci Adv ; 10(19): eadj9911, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728406

During cerebral cortex development, excitatory pyramidal neurons (PNs) establish specific projection patterns while receiving inputs from GABAergic inhibitory interneurons (INs). Whether these inhibitory inputs can shape PNs' projection patterns is, however, unknown. While layer 4 (L4) PNs of the primary somatosensory (S1) cortex are all born as long-range callosal projection neurons (CPNs), most of them acquire local connectivity upon activity-dependent elimination of their interhemispheric axons during postnatal development. Here, we demonstrate that precise developmental regulation of inhibition is key for the retraction of S1L4 PNs' callosal projections. Ablation of somatostatin INs leads to premature inhibition from parvalbumin INs onto S1L4 PNs and prevents them from acquiring their barrel-restricted local connectivity pattern. As a result, adult S1L4 PNs retain interhemispheric projections responding to tactile stimuli, and the mice lose whisker-based texture discrimination. Overall, we show that temporally ordered IN activity during development is key to shaping local ipsilateral S1L4 PNs' projection pattern, which is required for fine somatosensory processing.


GABAergic Neurons , Interneurons , Somatosensory Cortex , Animals , Interneurons/metabolism , Interneurons/physiology , Interneurons/cytology , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , GABAergic Neurons/cytology , Somatosensory Cortex/physiology , Somatosensory Cortex/metabolism , Somatosensory Cortex/cytology , Mice , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Parvalbumins/metabolism
2.
Addict Biol ; 29(5): e13403, 2024 May.
Article En | MEDLINE | ID: mdl-38735880

Synthetic opioids such as fentanyl contribute to the vast majority of opioid-related overdose deaths, but fentanyl use remains broadly understudied. Like other substances with misuse potential, opioids cause lasting molecular adaptations to brain reward circuits, including neurons in the ventral tegmental area (VTA). The VTA contains numerous cell types that play diverse roles in opioid use and relapse; however, it is unknown how fentanyl experience alters the transcriptional landscape in specific subtypes. Here, we performed single nuclei RNA sequencing to study transcriptional programs in fentanyl-experienced mice. Male and female C57/BL6 mice self-administered intravenous fentanyl (1.5 µg/kg/infusion) or saline for 10 days. After 24 h abstinence, VTA nuclei were isolated and prepared for sequencing on the 10× platform. We identified different patterns of gene expression across cell types. In dopamine neurons, we found enrichment of genes involved in growth hormone signalling. In dopamine-glutamate-GABA combinatorial neurons, and some GABA neurons, we found enrichment of genes involved in Pi3k-Akt signalling. In glutamate neurons, we found enrichment of genes involved in cholinergic signalling. We identified transcriptional regulators for the differentially expressed genes in each neuron cluster, including downregulated transcriptional repressor Bcl6, and upregulated transcription factor Tcf4. We also compared the fentanyl-induced gene expression changes identified in mouse VTA with a published rat dataset in bulk VTA, and found overlap in genes related to GABAergic signalling and extracellular matrix interaction. Together, we provide a comprehensive picture of how fentanyl self-administration alters the transcriptional landscape of the mouse VTA that serves as the foundation for future mechanistic studies.


Analgesics, Opioid , Fentanyl , Mice, Inbred C57BL , Ventral Tegmental Area , Animals , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Mice , Fentanyl/pharmacology , Male , Female , Analgesics, Opioid/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Self Administration , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Neurons/drug effects , Neurons/metabolism , Opioid-Related Disorders/genetics
3.
Acta Neuropathol ; 147(1): 80, 2024 May 07.
Article En | MEDLINE | ID: mdl-38714540

GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation-inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.


GABAergic Neurons , Interneurons , Tuberous Sclerosis , Interneurons/pathology , Interneurons/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis/metabolism , Humans , GABAergic Neurons/pathology , GABAergic Neurons/metabolism , Male , Female , Median Eminence/pathology , Median Eminence/metabolism , Somatostatin/metabolism , Child , Child, Preschool , Receptors, GABA-A/metabolism , Adolescent , Ganglionic Eminence
4.
J Mol Neurosci ; 74(2): 50, 2024 May 02.
Article En | MEDLINE | ID: mdl-38693434

Aneuploidy, having an aberrant genome, is gaining increasing attention in neurodegenerative diseases. It gives rise to proteotoxic stress as well as a stereotypical oxidative shift which makes these cells sensitive to internal and environmental stresses. A growing body of research from numerous laboratories suggests that many neurodegenerative disorders, especially Alzheimer's disease and frontotemporal dementia, are characterised by neuronal aneuploidy and the ensuing apoptosis, which may contribute to neuronal loss. Using Drosophila as a model, we investigated the effect of induced aneuploidy in GABAergic neurons. We found an increased proportion of aneuploidy due to Mad2 depletion in the third-instar larval brain and increased cell death. Depletion of Mad2 in GABAergic neurons also gave a defective climbing and seizure phenotype. Feeding animals an antioxidant rescued the climbing and seizure phenotype. These findings suggest that increased aneuploidy leads to higher oxidative stress in GABAergic neurons which causes cell death, climbing defects, and seizure phenotype. Antioxidant feeding represents a potential therapy to reduce the aneuploidy-driven neurological phenotype.


Aneuploidy , GABAergic Neurons , Oxidative Stress , Phenotype , Animals , GABAergic Neurons/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Seizures/genetics , Seizures/metabolism , Drosophila melanogaster/genetics , Brain/metabolism , Drosophila/genetics
5.
eNeuro ; 11(5)2024 May.
Article En | MEDLINE | ID: mdl-38658137

The primary motor cortex (M1) integrates sensory and cognitive inputs to generate voluntary movement. Its functional impairments have been implicated in the pathophysiology of motor symptoms in Parkinson's disease (PD). Specifically, dopaminergic degeneration and basal ganglia dysfunction entrain M1 neurons into the abnormally synchronized bursting pattern of activity throughout the cortico-basal ganglia-thalamocortical network. However, how degeneration of the midbrain dopaminergic neurons affects the anatomy, microcircuit connectivity, and function of the M1 network remains poorly understood. The present study examined whether and how the loss of dopamine (DA) affects the morphology, cellular excitability, and synaptic physiology of Layer 5 parvalbumin-expressing (PV+) cells in the M1 of mice of both sexes. Here, we reported that loss of midbrain dopaminergic neurons does not alter the number, morphology, and physiology of Layer 5 PV+ cells in M1. Moreover, we demonstrated that the number of perisomatic PV+ puncta of M1 pyramidal neurons as well as their functional innervation of cortical pyramidal neurons were not altered following the loss of DA. Together, the present study documents an intact GABAergic inhibitory network formed by PV+ cells following the loss of midbrain dopaminergic neurons.


Dopaminergic Neurons , Interneurons , Mesencephalon , Mice, Transgenic , Motor Cortex , Parvalbumins , Animals , Parvalbumins/metabolism , Motor Cortex/metabolism , Dopaminergic Neurons/metabolism , Interneurons/metabolism , Male , Female , Mesencephalon/metabolism , GABAergic Neurons/metabolism , Mice, Inbred C57BL , Mice , Neural Inhibition/physiology
6.
Nature ; 629(8011): 384-392, 2024 May.
Article En | MEDLINE | ID: mdl-38600385

Debate remains around the anatomical origins of specific brain cell subtypes and lineage relationships within the human forebrain1-7. Thus, direct observation in the mature human brain is critical for a complete understanding of its structural organization and cellular origins. Here we utilize brain mosaic variation within specific cell types as distinct indicators for clonal dynamics, denoted as cell-type-specific mosaic variant barcode analysis. From four hemispheres and two different human neurotypical donors, we identified 287 and 780 mosaic variants, respectively, that were used to deconvolve clonal dynamics. Clonal spread and allele fractions within the brain reveal that local hippocampal excitatory neurons are more lineage-restricted than resident neocortical excitatory neurons or resident basal ganglia GABAergic inhibitory neurons. Furthermore, simultaneous genome transcriptome analysis at both a cell-type-specific and a single-cell level suggests a dorsal neocortical origin for a subgroup of DLX1+ inhibitory neurons that disperse radially from an origin shared with excitatory neurons. Finally, the distribution of mosaic variants across 17 locations within one parietal lobe reveals that restriction of clonal spread in the anterior-posterior axis precedes restriction in the dorsal-ventral axis for both excitatory and inhibitory neurons. Thus, cell-type-resolved somatic mosaicism can uncover lineage relationships governing the development of the human forebrain.


Cell Lineage , GABAergic Neurons , Homeodomain Proteins , Mosaicism , Prosencephalon , Transcription Factors , Humans , Prosencephalon/cytology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cell Lineage/genetics , Male , Transcription Factors/metabolism , Transcription Factors/genetics , Neurons/cytology , Neurons/metabolism , Female , Hippocampus/cytology , Clone Cells/cytology , Clone Cells/metabolism , Single-Cell Analysis , Parietal Lobe/cytology , Alleles , Neocortex/cytology , Transcriptome
7.
J Physiol ; 602(10): 2343-2358, 2024 May.
Article En | MEDLINE | ID: mdl-38654583

Training rodents in a particularly difficult olfactory-discrimination (OD) task results in the acquisition of the ability to perform the task well, termed 'rule learning'. In addition to enhanced intrinsic excitability and synaptic excitation in piriform cortex pyramidal neurons, rule learning results in increased synaptic inhibition across the whole cortical network to the point where it precisely maintains the balance between inhibition and excitation. The mechanism underlying such precise inhibitory enhancement remains to be explored. Here, we use brain slices from transgenic mice (VGAT-ChR2-EYFP), enabling optogenetic stimulation of single GABAergic neurons and recordings of unitary synaptic events in pyramidal neurons. Quantal analysis revealed that learning-induced enhanced inhibition is mediated by increased quantal size of the evoked inhibitory events. Next, we examined the plasticity of synaptic inhibition induced by long-lasting, intrinsically evoked spike firing in post-synaptic neurons. Repetitive depolarizing current pulses from depolarized (-70 mV) or hyperpolarized (-90 mV) membrane potentials induced long-term depression (LTD) and long-term potentiation (LTP) of synaptic inhibition, respectively. We found a profound bidirectional increase in the ability to induce both LTD, mediated by L-type calcium channels, and LTP, mediated by R-type calcium channels after rule learning. Blocking the GABAB receptor reversed the effect of intrinsic stimulation at -90 mV from LTP to LTD. We suggest that learning greatly enhances the ability to modify the strength of synaptic inhibition of principal neurons in both directions. Such plasticity of synaptic plasticity allows fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule. KEY POINTS: Olfactory discrimination rule learning results in long-lasting enhancement of synaptic inhibition on piriform cortex pyramidal neurons. Quantal analysis of unitary inhibitory synaptic events, evoked by optogenetic minimal stimulation, revealed that enhanced synaptic inhibition is mediated by increased quantal size. Surprisingly, metaplasticity of synaptic inhibition, induced by intrinsically evoked repetitive spike firing, is increased bidirectionally. The susceptibility to both long-term depression (LTD) and long-term potentiation (LTP) of inhibition is enhanced after learning. LTD of synaptic inhibition is mediated by L-type calcium channels and LTP by R-type calcium channels. LTP is also dependent on activation of GABAB receptors. We suggest that learning-induced changes in the metaplasticity of synaptic inhibition enable the fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule.


Mice, Transgenic , Neuronal Plasticity , Pyramidal Cells , Animals , Neuronal Plasticity/physiology , Mice , Pyramidal Cells/physiology , GABAergic Neurons/physiology , Learning/physiology , Long-Term Potentiation/physiology , Male , Synapses/physiology , Optogenetics , Neural Inhibition/physiology , Piriform Cortex/physiology , Mice, Inbred C57BL , Long-Term Synaptic Depression/physiology
8.
Proc Natl Acad Sci U S A ; 121(16): e2317783121, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38588430

GABAergic inhibitory interneurons, originating from the embryonic ventral forebrain territories, traverse a convoluted migratory path to reach the neocortex. These interneuron precursors undergo sequential phases of tangential and radial migration before settling into specific laminae during differentiation. Here, we show that the developmental trajectory of FoxG1 expression is dynamically controlled in these interneuron precursors at critical junctures of migration. By utilizing mouse genetic strategies, we elucidate the pivotal role of precise changes in FoxG1 expression levels during interneuron specification and migration. Our findings underscore the gene dosage-dependent function of FoxG1, aligning with clinical observations of FOXG1 haploinsufficiency and duplication in syndromic forms of autism spectrum disorders. In conclusion, our results reveal the finely tuned developmental clock governing cortical interneuron development, driven by temporal dynamics and the dose-dependent actions of FoxG1.


Cerebral Cortex , Neocortex , Mice , Animals , Cerebral Cortex/metabolism , Cell Movement/physiology , Neurogenesis/physiology , Interneurons/physiology , Biomarkers/metabolism , GABAergic Neurons/physiology
9.
J Neurodev Disord ; 16(1): 21, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658850

BACKGROUND: Succinic semialdehyde dehydrogenase deficiency (SSADHD) represents a model neurometabolic disease at the fulcrum of translational research within the Boston Children's Hospital Intellectual and Developmental Disabilities Research Centers (IDDRC), including the NIH-sponsored natural history study of clinical, neurophysiological, neuroimaging, and molecular markers, patient-derived induced pluripotent stem cells (iPSC) characterization, and development of a murine model for tightly regulated, cell-specific gene therapy. METHODS: SSADHD subjects underwent clinical evaluations, neuropsychological assessments, biochemical quantification of γ-aminobutyrate (GABA) and related metabolites, electroencephalography (standard and high density), magnetoencephalography, transcranial magnetic stimulation, magnetic resonance imaging and spectroscopy, and genetic tests. This was parallel to laboratory molecular investigations of in vitro GABAergic neurons derived from induced human pluripotent stem cells (hiPSCs) of SSADHD subjects and biochemical analyses performed on a versatile murine model that uses an inducible and reversible rescue strategy allowing on-demand and cell-specific gene therapy. RESULTS: The 62 SSADHD subjects [53% females, median (IQR) age of 9.6 (5.4-14.5) years] included in the study had a reported symptom onset at ∼ 6 months and were diagnosed at a median age of 4 years. Language developmental delays were more prominent than motor. Autism, epilepsy, movement disorders, sleep disturbances, and various psychiatric behaviors constituted the core of the disorder's clinical phenotype. Lower clinical severity scores, indicating worst severity, coincided with older age (R= -0.302, p = 0.03), as well as age-adjusted lower values of plasma γ-aminobutyrate (GABA) (R = 0.337, p = 0.02) and γ-hydroxybutyrate (GHB) (R = 0.360, p = 0.05). While epilepsy and psychiatric behaviors increase in severity with age, communication abilities and motor function tend to improve. iPSCs, which were differentiated into GABAergic neurons, represent the first in vitro neuronal model of SSADHD and express the neuronal marker microtubule-associated protein 2 (MAP2), as well as GABA. GABA-metabolism in induced GABAergic neurons could be reversed using CRISPR correction of the pathogenic variants or mRNA transfection and SSADHD iPSCs were associated with excessive glutamatergic activity and related synaptic excitation. CONCLUSIONS: Findings from the SSADHD Natural History Study converge with iPSC and animal model work focused on a common disorder within our IDDRC, deepening our knowledge of the pathophysiology and longitudinal clinical course of a complex neurodevelopmental disorder. This further enables the identification of biomarkers and changes throughout development that will be essential for upcoming targeted trials of enzyme replacement and gene therapy.


Amino Acid Metabolism, Inborn Errors , Developmental Disabilities , Induced Pluripotent Stem Cells , Succinate-Semialdehyde Dehydrogenase , Adolescent , Animals , Child , Child, Preschool , Female , Humans , Male , Mice , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/metabolism , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , GABAergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics , Succinate-Semialdehyde Dehydrogenase/deficiency , Succinate-Semialdehyde Dehydrogenase/metabolism , Succinate-Semialdehyde Dehydrogenase/genetics
10.
Nat Commun ; 15(1): 2884, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570488

Increasing evidence has revealed the large-scale nonstationary synchronizations as traveling waves in spontaneous neural activity. However, the interplay of various cell types in fine-tuning these spatiotemporal patters remains unclear. Here, we performed comprehensive exploration of spatiotemporal synchronizing structures across different cell types, states (awake, anesthesia, motion) and developmental axis in male mice. We found traveling waves in glutamatergic neurons exhibited greater variety than those in GABAergic neurons. Moreover, the synchronizing structures of GABAergic neurons converged toward those of glutamatergic neurons during development, but the evolution of waves exhibited varying timelines for different sub-type interneurons. Functional connectivity arises from both standing and traveling waves, and negative connections can be elucidated by the spatial propagation of waves. In addition, some traveling waves were correlated with the spatial distribution of gene expression. Our findings offer further insights into the neural underpinnings of traveling waves, functional connectivity, and resting-state networks, with cell-type specificity and developmental perspectives.


GABAergic Neurons , Male , Mice , Animals
11.
Nat Commun ; 15(1): 2891, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570514

Animals are inherently motivated to explore social novelty cues over familiar ones, resulting in a novelty preference (NP), although the behavioral and circuit bases underlying NP are unclear. Combining calcium and neurotransmitter sensors with fiber photometry and optogenetics in mice, we find that mesolimbic dopamine (DA) neurotransmission is strongly and predominantly activated by social novelty controlling bout length of interaction during NP, a response significantly reduced by familiarity. In contrast, interpeduncular nucleus (IPN) GABAergic neurons that project to the lateral dorsal tegmentum (LDTg) were inhibited by social novelty but activated during terminations with familiar social stimuli. Inhibition of this pathway during NP increased interaction and bout length with familiar social stimuli, while activation reduced interaction and bout length with novel social stimuli via decreasing DA neurotransmission. These data indicate interest towards novel social stimuli is encoded by mesolimbic DA which is dynamically regulated by an IPN→LDTg circuit to control NP.


Dopamine , Interpeduncular Nucleus , Mice , Animals , Dopamine/metabolism , Tegmentum Mesencephali/metabolism , Interpeduncular Nucleus/metabolism , Synaptic Transmission , GABAergic Neurons/metabolism
12.
Transl Psychiatry ; 14(1): 197, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38670959

Alcohol use and anxiety disorders occur in both males and females, but despite sharing similar presentation and classical symptoms, the prevalence of alcohol use disorder (AUD) is lower in females. While anxiety is a symptom and comorbidity shared by both sexes, the common underlying mechanism that leads to AUD and the subsequent development of anxiety is still understudied. Using a rodent model of adolescent intermittent ethanol (AIE) exposure in both sexes, we investigated the epigenetic mechanism mediated by enhancer of zeste 2 (EZH2), a histone methyltransferase, in regulating both the expression of activity-regulated cytoskeleton-associated protein (Arc) and an anxiety-like phenotype in adulthood. Here, we report that EZH2 protein levels were significantly higher in PKC-δ positive GABAergic neurons in the central nucleus of amygdala (CeA) of adult male and female rats after AIE. Reducing protein and mRNA levels of EZH2 using siRNA infusion in the CeA prevented AIE-induced anxiety-like behavior, increased H3K27me3, decreased H3K27ac at the Arc synaptic activity response element (SARE) site, and restored deficits in Arc mRNA and protein expression in both male and female adult rats. Our data indicate that an EZH2-mediated epigenetic mechanism in the CeA plays an important role in regulating anxiety-like behavior and Arc expression after AIE in both male and female rats in adulthood. This study suggests that EZH2 may serve as a tractable drug target for the treatment of adult psychopathology after adolescent alcohol exposure.


Anxiety , Central Amygdaloid Nucleus , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Ethanol , Animals , Male , Female , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Rats , Anxiety/metabolism , Anxiety/genetics , Ethanol/pharmacology , Disease Models, Animal , Alcoholism/genetics , Alcoholism/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Rats, Sprague-Dawley , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
13.
PLoS One ; 19(4): e0300544, 2024.
Article En | MEDLINE | ID: mdl-38656972

Obesity is a major global health epidemic that has adverse effects on both the people affected as well as the cost to society. Several anti-obesity drugs that target GLP-1 receptors have recently come to the market. Here, we describe the effects of tesofensine, a novel anti-obesity drug that acts as a triple monoamine neurotransmitter reuptake inhibitor. Using various techniques, we investigated its effects on weight loss and underlying neuronal mechanisms in mice and rats. These include behavioral tasks, DeepLabCut videotaped analysis, electrophysiological ensemble recordings, optogenetic activation, and chemogenetic silencing of GABAergic neurons in the Lateral Hypothalamus (LH). We found that tesofensine induces a greater weight loss in obese rats than lean rats, while differentially modulating the neuronal ensembles and population activity in LH. In Vgat-ChR2 and Vgat-IRES-cre transgenic mice, we found for the first time that tesofensine inhibited a subset of LH GABAergic neurons, reducing their ability to promote feeding behavior, and chemogenetically silencing them enhanced tesofensine's food-suppressing effects. Unlike phentermine, a dopaminergic appetite suppressant, tesofensine causes few, if any, head-weaving stereotypy at therapeutic doses. Most importantly, we found that tesofensine prolonged the weight loss induced by 5-HTP, a serotonin precursor, and blocked the body weight rebound that often occurs after weight loss. Behavioral studies on rats with the tastant sucrose indicated that tesofensine's appetite suppressant effects are independent of taste aversion and do not directly affect the perception of sweetness or palatability of sucrose. In summary, our data provide new insights into the effects of tesofensine on weight loss and the underlying neuronal mechanisms, suggesting that tesofensine may be an effective treatment for obesity and that it may be a valuable adjunct to other appetite suppressants to prevent body weight rebound.


Anti-Obesity Agents , Bridged Bicyclo Compounds, Heterocyclic , GABAergic Neurons , Obesity , Animals , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Rats , Mice , Anti-Obesity Agents/pharmacology , Male , Obesity/drug therapy , Obesity/metabolism , Feeding Behavior/drug effects , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice, Transgenic , Weight Loss/drug effects , Rats, Sprague-Dawley
14.
J Neurosci ; 44(19)2024 May 08.
Article En | MEDLINE | ID: mdl-38565288

Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.


Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Mice, Inbred C57BL , Signal Transduction , TRPM Cation Channels , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Mice , TRPM Cation Channels/metabolism , Signal Transduction/physiology , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Receptors, GABA-A/metabolism , Hippocampus/metabolism , Neural Inhibition/physiology , GABAergic Neurons/metabolism , Heart Arrest/complications , Heart Arrest/metabolism
15.
Neurosci Biobehav Rev ; 161: 105651, 2024 Jun.
Article En | MEDLINE | ID: mdl-38579901

GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.


Brain , gamma-Aminobutyric Acid , gamma-Aminobutyric Acid/metabolism , Humans , Animals , Brain/growth & development , Brain/metabolism , Receptors, GABA-A/metabolism , GABAergic Neurons/physiology , GABAergic Neurons/metabolism
16.
Nat Neurosci ; 27(5): 862-872, 2024 May.
Article En | MEDLINE | ID: mdl-38528203

The mammalian telencephalon contains distinct GABAergic projection neuron and interneuron types, originating in the germinal zone of the embryonic basal ganglia. How genetic information in the germinal zone determines cell types is unclear. Here we use a combination of in vivo CRISPR perturbation, lineage tracing and ChIP-sequencing analyses and show that the transcription factor MEIS2 favors the development of projection neurons by binding enhancer regions in projection-neuron-specific genes during mouse embryonic development. MEIS2 requires the presence of the homeodomain transcription factor DLX5 to direct its functional activity toward the appropriate binding sites. In interneuron precursors, the transcription factor LHX6 represses the MEIS2-DLX5-dependent activation of projection-neuron-specific enhancers. Mutations of Meis2 result in decreased activation of regulatory enhancers, affecting GABAergic differentiation. We propose a differential binding model where the binding of transcription factors at cis-regulatory elements determines differential gene expression programs regulating cell fate specification in the mouse ganglionic eminence.


Embryonic Development , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Transcription Factors , Animals , Mice , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Embryonic Development/physiology , Enhancer Elements, Genetic/genetics , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Cell Differentiation/physiology , Interneurons/metabolism , Interneurons/physiology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Neurogenesis/physiology , Nerve Tissue Proteins
17.
J Physiol ; 602(8): 1733-1757, 2024 Apr.
Article En | MEDLINE | ID: mdl-38493320

Differentiating between auditory signals of various emotional significance plays a crucial role in an individual's ability to thrive and excel in social interactions and in survival. Multiple approaches, including anatomical studies, electrophysiological investigations, imaging techniques, optogenetics and chemogenetics, have confirmed that the auditory cortex (AC) impacts fear-related behaviours driven by auditory stimuli by conveying auditory information to the lateral amygdala (LA) through long-range excitatory glutamatergic and GABAergic connections. In addition, the LA provides glutamatergic projections to the AC which are important to fear memory expression and are modified by associative fear learning. Here we test the hypothesis that the LA also sends long-range direct inhibitory inputs to the cortex. To address this fundamental question, we used anatomical and electrophysiological approaches, allowing us to directly assess the nature of GABAergic inputs from the LA to the AC in the mouse. Our findings elucidate the existence of a long-range inhibitory pathway from the LA to the AC (LAC) via parvalbumin-expressing (LAC-Parv) and somatostatin-expressing (LAC-SOM) neurons. This research identifies distinct electrophysiological properties for genetically defined long-range GABAergic neurons involved in the communication between the LA and the cortex (LAC-Parv inhibitory projections → AC neurons; LAC-Som inhibitory projections → AC neurons) within the lateral amygdala cortical network. KEY POINTS: The mouse auditory cortex receives inputs from the lateral amygdala. Retrograde viral tracing techniques allowed us to identify two previously undescribed lateral amygdala to auditory cortex (LAC) GABAergic projecting neurons. Extensive electrophysiological, morphological and anatomical characterization of LAC neurons is provided here, demonstrating key differences in the three populations. This study paves the way for a better understanding of the growing complexity of the cortico-amygdala-cortico circuit.


Auditory Cortex , Mice , Animals , Auditory Cortex/physiology , Amygdala/physiology , GABAergic Neurons/physiology , Parvalbumins/metabolism
18.
Cell Mol Life Sci ; 81(1): 147, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38502309

GABAergic interneurons are poised with the capacity to shape circuit output via inhibitory gating. How early in the development of medial vestibular nucleus (MVN) are GABAergic neurons recruited for feedforward shaping of outputs to higher centers for spatial navigation? The role of early GABAergic transmission in assembling vestibular circuits for spatial navigation was explored by neonatal perturbation. Immunohistochemistry and confocal imaging were utilized to reveal the expression of parvalbumin (PV)-expressing MVN neurons and their perineuronal nets. Whole-cell patch-clamp recording, coupled with optogenetics, was conducted in vitro to examine the synaptic function of MVN circuitry. Chemogenetic targeting strategy was also employed in vivo to manipulate neuronal activity during navigational tests. We found in rats a neonatal critical period before postnatal day (P) 8 in which competitive antagonization of GABAergic transmission in the MVN retarded maturation of inhibitory neurotransmission, as evidenced by deranged developmental trajectory for excitation/inhibition ratio and an extended period of critical period-like plasticity in GABAergic transmission. Despite increased number of PV-expressing GABAergic interneurons in the MVN, optogenetic-coupled patch-clamp recording indicated null-recruitment of these neurons in tuning outputs along the ascending vestibular pathway. Such perturbation not only offset output dynamics of ascending MVN output neurons, but was further accompanied by impaired vestibular-dependent navigation in adulthood. The same perturbations were however non-consequential when applied after P8. Results highlight neonatal GABAergic transmission as key to establishing feedforward output dynamics to higher brain centers for spatial cognition and navigation.


Spatial Navigation , Rats , Animals , Interneurons , Synaptic Transmission , Vestibular Nuclei/metabolism , GABAergic Neurons
19.
Commun Biol ; 7(1): 339, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38503808

The neural circuits underlying sleep-wakefulness and general anesthesia have not been fully investigated. The GABAergic neurons in the bed nucleus of the stria terminalis (BNST) play a critical role in stress and fear that relied on heightened arousal. Nevertheless, it remains unclear whether BNST GABAergic neurons are involved in the regulation of sleep-wakefulness and anesthesia. Here, using in vivo fiber photometry combined with electroencephalography, electromyography, and video recordings, we found that BNST GABAergic neurons exhibited arousal-state-dependent alterations, with high activities in both wakefulness and rapid-eye movement sleep, but suppressed during anesthesia. Optogenetic activation of these neurons could initiate and maintain wakefulness, and even induce arousal from anesthesia. However, chronic lesion of BNST GABAergic neurons altered spontaneous sleep-wakefulness architecture during the dark phase, but not induction and emergence from anesthesia. Furthermore, we also discovered that the BNST-ventral tegmental area pathway might participate in promoting wakefulness and reanimation from steady-state anesthesia. Collectively, our study explores new elements in neural circuit mechanisms underlying sleep-wakefulness and anesthesia, which may contribute to a more comprehensive understanding of consciousness and the development of innovative anesthetics.


Septal Nuclei , Wakefulness , Wakefulness/physiology , Septal Nuclei/physiology , Sleep/physiology , GABAergic Neurons/physiology , Anesthesia, General
20.
Elife ; 122024 Mar 21.
Article En | MEDLINE | ID: mdl-38512339

Although the midbrain dopamine (DA) system plays a crucial role in higher cognitive functions, including updating and maintaining short-term memory, the encoding properties of the somatic spiking activity of ventral tegmental area (VTA) DA neurons for short-term memory computations have not yet been identified. Here, we probed and analyzed the activity of optogenetically identified DA and GABA neurons while mice engaged in short-term memory-dependent behavior in a T-maze task. Single-neuron analysis revealed that significant subpopulations of DA and GABA neurons responded differently between left and right trials in the memory delay. With a series of control behavioral tasks and regression analysis tools, we show that firing rate differences are linked to short-term memory-dependent decisions and cannot be explained by reward-related processes, motivated behavior, or motor-related activities. This evidence provides novel insights into the mnemonic encoding activities of midbrain DA and GABA neurons.


Dopamine , Ventral Tegmental Area , Animals , Mice , GABAergic Neurons , Cognition , Memory, Short-Term
...