Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.319
Filter
1.
bioRxiv ; 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39257744

ABSTRACT

Dystroglycan is a cell adhesion molecule that localizes to synapses throughout the nervous system. While Dystroglycan is required to maintain inhibitory synapses from cerebellar molecular layer interneurons (MLIs) onto Purkinje cells (PCs) whether initial synaptogenesis during development is dependent on Dystroglycan has not been examined. We show that conditional deletion of Dystroglycan from Purkinje cells prior to synaptogenesis results in impaired MLI:PC synapse formation and function due to reduced presynaptic inputs and abnormal postsynaptic GABAA receptor clustering. Using genetic manipulations that disrupt glycosylation of Dystroglycan or truncate its cytoplasmic domain, we show that Dystroglycan's role in synapse function requires both extracellular and intracellular interactions, whereas synapse formation requires only extracellular interactions. Together, these findings provide molecular insight into the mechanism of inhibitory synapse formation and maintenance in cerebellar cortex.

2.
Geroscience ; 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39343864

ABSTRACT

Changes to neuronal connectivity are believed to be a key factor in cognitive impairments associated with normal aging. Because of its effect on activities of daily living, deficient motor control is a critical type of cognitive decline to understand. Diminished inhibitory networks in the cortex are implicated in such motor control deficits, pointing to the connectivity of inhibitory cortical interneurons as an important area for study. Here, we used chronic two-photon microscopy to track the structural plasticity of en passant boutons (EPBs) of parvalbumin-positive interneurons in the mouse motor cortex in the first longitudinal, in vivo study of inhibitory interneuron synapses in the context of aging. Young (3-5 months) and aged (23-28 months) mice underwent training on the accelerating rotarod to evoke motor learning-induced structural plasticity. Our analysis reveals that, in comparison with axons from young mice, those from aged mice have fewer EPBs at baseline that also tend to be larger in size. Aged axons also express learning-related structural plasticity-like new bouton stabilization and bouton enlargement-that is less persistent than that of young axons. This study reveals striking baseline differences in young and aged axon morphology as well as differences in the deployment of learning-related structural plasticity across axons.

3.
BMC Neurosci ; 25(1): 45, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333878

ABSTRACT

BACKGROUND: Exposure to chemical toxins, including insecticides, harms bodily organs like the brain. This study examined the neuroprotective of thymoquinone on the cypermethrin's harmful effects on the histoarchitecture of the dentate gyrus and motor deficit in the dentate gyrus. METHODS: Forty adult male rats (180-200 g) were randomly divided into 5 groups (n = 8 per group). Groups I, II, III, IV, and V received oral administration of 0.5 ml of phosphate-buffered saline, cypermethrin (20 mg/kg), thymoquinone (10 mg/kg), cypermethrin (20 mg/kg) + thymoquinone (5 mg/kg), and cypermethrin (20 mg/kg) + thymoquinone (10 mg/kg) for 14 days respectively. The novel object recognition test that assesses intermediate-term memory was done on days 14 and 21 of the experiment. At the end of these treatments, the animals were euthanized and taken for cytoarchitectural (hematoxylin and eosin; Cresyl violet) and immunohistochemical studies (Nuclear factor erythroid 2-related factor 2 (Nrf2), Parvalbumin, and B-cell lymphoma 2 (Bcl2). RESULT: The study shows that thymoquinone at 5 and 10 mg/kg improved Novelty preference and discrimination index. Thymoquinone enhanced Nissl body integrity, increased GABBAergic interneuron expression, nuclear factor erythroid 2-derived factor 2, and enhanced Bcl-2 expression in the dentate gyrus. It also improved the concentration of nuclear factor erythroid 2-derived factor 2, increased the activities of superoxide dismutase and glutathione, and decreased the concentration of malondialdehyde level against cypermethrin-induced neurotoxicity. CONCLUSION: thymoquinone could be a therapeutic agent against cypermethrin poisoning.


Subject(s)
Benzoquinones , Dentate Gyrus , GABAergic Neurons , Memory Disorders , NF-E2-Related Factor 2 , Oxidative Stress , Pyrethrins , Signal Transduction , Animals , Pyrethrins/toxicity , Male , Oxidative Stress/drug effects , Benzoquinones/pharmacology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Signal Transduction/drug effects , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Rats , NF-E2-Related Factor 2/metabolism , Insecticides/toxicity , Neuroprotective Agents/pharmacology , Rats, Wistar
4.
J Physiol ; 602(17): 4195-4213, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39141819

ABSTRACT

The subiculum is a key region of the brain involved in the initiation of pathological activity in temporal lobe epilepsy, and local GABAergic inhibition is essential to prevent subicular-originated epileptiform discharges. Subicular pyramidal cells may be easily distinguished into two classes based on their different firing patterns. Here, we have compared the strength of the GABAa receptor-mediated inhibitory postsynaptic currents received by regular- vs. burst-firing subicular neurons and their dynamic modulation by the activation of µ opioid receptors. We have taken advantage of the sequential re-patching of the same cell to initially classify pyramidal neurons according to their firing patters, and then to measure GABAergic events triggered by the optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons. Activation of parvalbumin-expressing cells generated larger responses in postsynaptic burst-firing neurons whereas the opposite was observed for currents evoked by the stimulation of somatostatin-expressing interneurons. In all cases, events depended critically on ω-agatoxin IVA- but not on ω-conotoxin GVIA-sensitive calcium channels. Optogenetic GABAergic input originating from both parvalbumin- and somatostatin-expressing cells was reduced in amplitude following the exposure to a µ opioid receptor agonist. The kinetics of this pharmacological sensitivity was different in regular- vs. burst-firing neurons, but only when responses were evoked by the activation of parvalbumin-expressing neurons, whereas no differences were observed when somatostatin-expressing cells were stimulated. In conclusion, our results show that a high degree of complexity regulates the organizing principles of subicular GABAergic inhibition, with the interaction of pre- and postsynaptic diversity at multiple levels. KEY POINTS: Optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons (PVs and SOMs) triggers inhibitory postsynaptic currents (IPSCs) in both regular- and burst-firing (RFs and BFs) subicular pyramidal cells. The amplitude of optogenetically evoked IPSCs from PVs (PV-opto IPSCs) is larger in BFs whereas IPSCs generated by the light activation of SOMs (SOM-opto IPSCs) are larger in RFs. Both PV- and SOM-opto IPSCs critically depend on ω-agatoxin IVA-sensitive P/Q type voltage-gated calcium channels, whereas no major effects are observed following exposure to ω-conotoxin GVIA, suggesting no significant involvement of N-type channels. The amplitude of both PV- and SOM-opto IPSCs is reduced by the probable pharmacological activation of presynaptic µ opioid receptors, with a faster kinetics of the effect observed in PV-opto IPSCs from RFs vs. BFs, but not in SOM-opto IPSCs. These results help us understand the complex interactions between different layers of diversity regulating GABAergic input onto subicular microcircuits.


Subject(s)
Parvalbumins , Pyramidal Cells , Somatostatin , Animals , Pyramidal Cells/physiology , Mice , Somatostatin/metabolism , Parvalbumins/metabolism , Interneurons/physiology , Inhibitory Postsynaptic Potentials , Male , GABAergic Neurons/physiology , GABAergic Neurons/metabolism , Hippocampus/physiology , Hippocampus/cytology , Optogenetics , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/physiology , Mice, Inbred C57BL , Female , Receptors, GABA-A/metabolism , Receptors, GABA-A/physiology
5.
bioRxiv ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39131281

ABSTRACT

Prior reward is a potent cue for attentional capture, but the underlying neurobiology is largely unknown. In a novel whisker touch detection task, we show that mice flexibly shift attention between specific whiskers on a trial-by-trial timescale, guided by the recent history of stimulus-reward association. Two-photon calcium imaging and spike recordings revealed a robust neurobiological correlate of attention in the somatosensory cortex (S1), boosting sensory responses to the attended whisker in L2/3 and L5, but not L4. Attentional boosting in L2/3 pyramidal cells was topographically precise and whisker-specific, and shifted receptive fields toward the attended whisker. L2/3 VIP interneurons were broadly activated by whisker stimuli, motion, and arousal but did not carry a whisker-specific attentional signal, and thus did not mediate spatially focused tactile attention. Together, these findings establish a new model of focal attention in the mouse whisker tactile system, showing that the history of stimuli and rewards in the recent past can dynamically engage local modulation in cortical sensory maps to guide flexible shifts in ongoing behavior.

6.
J Neurophysiol ; 132(2): 403-417, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39106208

ABSTRACT

Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum (DS), ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared with males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared with males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared with other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.NEW & NOTEWORTHY Cholinergic interneurons (ChIs) within the dorsal and ventral striatum can exert a major influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum, specifically the nucleus accumbens (NAc) shell, are understudied. Here, we report that NAc shell ChIs have heterogeneous ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide corticotropin releasing factor (CRF) and by the estrous cycle.


Subject(s)
Cholinergic Neurons , Corticotropin-Releasing Hormone , Interneurons , Nucleus Accumbens , Animals , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Female , Male , Interneurons/physiology , Interneurons/metabolism , Nucleus Accumbens/physiology , Nucleus Accumbens/metabolism , Nucleus Accumbens/cytology , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , Estrous Cycle/physiology , Action Potentials/physiology , Mice
7.
Cell Rep ; 43(9): 114659, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39180750

ABSTRACT

Empathy, crucial for social interaction, is impaired across various neuropsychiatric conditions. However, the genetic and neural underpinnings of empathy variability remain elusive. By combining forward genetic mapping with transcriptome analysis, we discover that aryl hydrocarbon receptor nuclear translocator 2 (ARNT2) is a key driver modulating observational fear, a basic form of affective empathy. Disrupted ARNT2 expression in the anterior cingulate cortex (ACC) reduces affect sharing in mice. Specifically, selective ARNT2 ablation in somatostatin (SST)-expressing interneurons leads to decreased pyramidal cell excitability, increased spontaneous firing, aberrant Ca2+ dynamics, and disrupted theta oscillations in the ACC, resulting in reduced vicarious freezing. We further demonstrate that ARNT2-expressing SST interneurons govern affective state discrimination, uncovering a potential mechanism by which ARNT2 polymorphisms associate with emotion recognition in humans. Our findings advance our understanding of the molecular mechanism controlling empathic capacity and highlight the neural substrates underlying social affective dysfunctions in psychiatric disorders.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator , Empathy , Interneurons , Prefrontal Cortex , Somatostatin , Animals , Empathy/physiology , Mice , Interneurons/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Somatostatin/metabolism , Male , Prefrontal Cortex/metabolism , Humans , Gyrus Cinguli/metabolism , Mice, Inbred C57BL , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Pyramidal Cells/metabolism , Female
8.
Front Neural Circuits ; 18: 1436915, 2024.
Article in English | MEDLINE | ID: mdl-39091993

ABSTRACT

We provide a brief (and unabashedly biased) overview of the pre-transcriptomic history of somatostatin interneuron taxonomy, followed by a chronological summary of the large-scale, NIH-supported effort over the last ten years to generate a comprehensive, single-cell RNA-seq-based taxonomy of cortical neurons. Focusing on somatostatin interneurons, we present the perspective of experimental neuroscientists trying to incorporate the new classification schemes into their own research while struggling to keep up with the ever-increasing number of proposed cell types, which seems to double every two years. We suggest that for experimental analysis, the most useful taxonomic level is the subdivision of somatostatin interneurons into ten or so "supertypes," which closely agrees with their more traditional classification by morphological, electrophysiological and neurochemical features. We argue that finer subdivisions ("t-types" or "clusters"), based on slight variations in gene expression profiles but lacking clear phenotypic differences, are less useful to researchers and may actually defeat the purpose of classifying neurons to begin with. We end by stressing the need for generating novel tools (mouse lines, viral vectors) for genetically targeting distinct supertypes for expression of fluorescent reporters, calcium sensors and excitatory or inhibitory opsins, allowing neuroscientists to chart the input and output synaptic connections of each proposed subtype, reveal the position they occupy in the cortical network and examine experimentally their roles in sensorimotor behaviors and cognitive brain functions.


Subject(s)
Interneurons , Somatostatin , Animals , Somatostatin/metabolism , Interneurons/classification , Interneurons/physiology , Interneurons/metabolism , Interneurons/cytology , Humans
9.
Hippocampus ; 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138952

ABSTRACT

The processing of rich synaptic information in the dentate gyrus (DG) relies on a diverse population of inhibitory GABAergic interneurons to regulate cellular and circuit activity, in a layer-specific manner. Metabotropic GABAB-receptors (GABABRs) provide powerful inhibition to the DG circuit, on timescales consistent with behavior and learning, but their role in controlling the activity of interneurons is poorly understood with respect to identified cell types. We hypothesize that GABABRs display cell type-specific heterogeneity in signaling strength, which will have direct ramifications for signal processing in DG networks. To test this, we perform in vitro whole-cell patch-clamp recordings from identified DG principal cells and interneurons, followed by GABABR pharmacology, photolysis of caged GABA, and extracellular stimulation of endogenous GABA release to classify the cell type-specific inhibitory potential. Based on our previous classification of DG interneurons, we show that postsynaptic GABABR-mediated currents are present on all interneuron types albeit at different amplitudes, dependent largely on soma location and synaptic targets. GABABRs were coupled to inwardly-rectifying K+ channels that strongly reduced the excitability of those interneurons where large currents were observed. These data provide a systematic characterization of GABABR signaling in the rat DG to provide greater insight into circuit dynamics.

10.
Biol Psychiatry Glob Open Sci ; 4(5): 100340, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140003

ABSTRACT

Background: The renin-angiotensin system has been identified as a potential therapeutic target for posttraumatic stress disorder, although its mechanisms are not well understood. Brain angiotensin type 2 receptors (AT2Rs) are a subtype of angiotensin II receptors located in stress and anxiety-related regions, including the medial prefrontal cortex (mPFC), but their function and mechanism in the mPFC remain unexplored. Therefore, we used a combination of imaging, cre/lox, and behavioral methods to investigate mPFC-AT2R-expressing neurons in fear and stess related behavior. Methods: To characterize mPFC-AT2R-expressing neurons in the mPFC, AT2R-Cre/tdTomato male and female mice were used for immunohistochemistry. mPFC brain sections were stained with glutamatergic or interneuron markers, and density of AT2R+ cells and colocalization with each marker were quantified. To assess fear-related behaviors in AT2R-flox mice, we selectively deleted AT2R from mPFC neurons using a Cre-expressing adeno-associated virus. Mice then underwent Pavlovian auditory fear conditioning, elevated plus maze, and open field testing. Results: Immunohistochemistry results revealed that AT2R was densely expressed throughout the mPFC and primarily expressed in somatostatin interneurons in a sex-dependent manner. Following fear conditioning, mPFC-AT2R Cre-lox deletion impaired extinction and increased exploratory behavior in female but not male mice, while locomotion was unaltered by mPFC-AT2R deletion in both sexes. Conclusions: These results identify mPFC-AT2R+ neurons as a novel subgroup of somatostatin interneurons and reveal their role in regulating fear learning in a sex-dependent manner, potentially offering insights into novel therapeutic targets for posttraumatic stress disorder.


Posttraumatic stress disorder (PTSD) is a significant predictor of cardiovascular disease (CVD), although the underlying mechanisms are poorly understood. The brain renin-angiotensin system (RAS) is important for cardiovascular and emotional stress regulation and may better help understand the link between PTSD and CVD risk. Our research reveals that the brain angiotensin II type 2 receptor (AT2R) subtype is located on specific somatostatin (SOM+) interneurons in the medial prefrontal cortex (mPFC) and plays a role in fear memory extinction, particularly in females. These findings reveal a role for the mPFC-AT2R in fear-based learning and memory, offering potential insights into the mechanisms underlying the PTSD-CVD association and therapeutic strategies.

11.
bioRxiv ; 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39149275

ABSTRACT

Social memory impairments in Mecp2 knockout (KO) mice result from altered neuronal activity in the monosynaptic projection from the ventral hippocampus (vHIP) to the medial prefrontal cortex (mPFC). The hippocampal network is hyperactive in this model for Rett syndrome, and such atypically heightened neuronal activity propagates to the mPFC through this monosynaptic projection, resulting in altered mPFC network activity and social memory deficits. However, the underlying mechanism of cellular dysfunction within this projection between vHIP pyramidal neurons (PYR) and mPFC PYRs and parvalbumin interneurons (PV-IN) resulting in social memory impairments in Mecp2 KO mice has yet to be elucidated. We confirmed social memory (but not sociability) deficits in Mecp2 KO mice using a new 4-chamber social memory arena, designed to minimize the impact of the tethering to optical fibers required for simultaneous in vivo fiber photometry of Ca2+-sensor signals during social interactions. mPFC PYRs of wildtype (WT) mice showed increases in Ca2+ signal amplitude during explorations of a novel toy mouse and interactions with both familiar and novel mice, while PYRs of Mecp2 KO mice showed smaller Ca2+ signals during interactions only with live mice. On the other hand, mPFC PV-INs of Mecp2 KO mice showed larger Ca2+ signals during interactions with a familiar cage-mate compared to those signals in PYRs, a difference absent in the WT mice. These observations suggest atypically heightened inhibition and impaired excitation in the mPFC network of Mecp2 KO mice during social interactions, potentially driving their deficit in social memory.

12.
Elife ; 132024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012795

ABSTRACT

Axo-axonic cells (AACs), also called chandelier cells (ChCs) in the cerebral cortex, are the most distinctive type of GABAergic interneurons described in the neocortex, hippocampus, and basolateral amygdala (BLA). AACs selectively innervate glutamatergic projection neurons (PNs) at their axon initial segment (AIS), thus may exert decisive control over PN spiking and regulate PN functional ensembles. However, the brain-wide distribution, synaptic connectivity, and circuit function of AACs remain poorly understood, largely due to the lack of specific and reliable experimental tools. Here, we have established an intersectional genetic strategy that achieves specific and comprehensive targeting of AACs throughout the mouse brain based on their lineage (Nkx2.1) and molecular (Unc5b, Pthlh) markers. We discovered that AACs are deployed across essentially all the pallium-derived brain structures, including not only the dorsal pallium-derived neocortex and medial pallium-derived hippocampal formation, but also the lateral pallium-derived claustrum-insular complex, and the ventral pallium-derived extended amygdaloid complex and olfactory centers. AACs are also abundant in anterior olfactory nucleus, taenia tecta, and lateral septum. AACs show characteristic variations in density across neocortical areas and layers and across subregions of the hippocampal formation. Neocortical AACs comprise multiple laminar subtypes with distinct dendritic and axonal arborization patterns. Retrograde monosynaptic tracing from AACs across neocortical, hippocampal, and BLA regions reveal shared as well as distinct patterns of synaptic input. Specific and comprehensive targeting of AACs facilitates the study of their developmental genetic program and circuit function across brain structures, providing a ground truth platform for understanding the conservation and variation of a bona fide cell type across brain regions and species.


Whether we are memorising facts or reacting to a loud noise, nerve cells in different brain areas must be able to communicate with one another through precise, meaningful signals. Specialized nerve cells known as interneurons act as "traffic lights" to precisely regulate when and where this information flows in neural circuits. Axo-axonic cells are a rare type of inhibitory interneuron that are thought to be particularly important for controlling the passage of information between different groups of excitatory neurons. This is because they only connect to one key part of their target cell ­ the axon-initial segment ­ where the electrical signals needed for brain communication (known as action potentials) are initiated. Since axo-axonic cells are inhibitory interneurons, this connection effectively allows them to 'veto' the generation of these signals at their source. Although axo-axonic cells have been identified in three brain regions using traditional anatomical methods, there were no 'tags' readily available that can reliably identify them. Therefore, much about these cells remained unknown, including how widespread they are in the mammalian brain. To solve this problem, Raudales et al. investigated which genes are switched on in axo-axonic cells but not in other cells, identifying a unique molecular signature that could be used to mark, record, and manipulate these cells. Microscopy imaging of brain tissue from mice in which axo-axonic cells had been identified revealed that they are present in many more brain areas than previously thought, including nearly all regions of the broadly defined cerebral cortex and even the hypothalamus, which controls many innate behaviors. Axo-axonic cells were also 'wired up' differently, depending on where they were located; for example, those in brain areas associated with memory and emotions had wider-ranging input connections than other areas. The finding of Raudales et al. provide, for the first time, a method to directly track and manipulate axo-axonic cells in the brain. Since dysfunction in axo-axonic cells is also associated with neurological disorders like epilepsy and schizophrenia, gaining an insight into their distribution and connectivity could help to develop better treatments for these conditions.


Subject(s)
GABAergic Neurons , Interneurons , Animals , Interneurons/physiology , Interneurons/metabolism , GABAergic Neurons/physiology , GABAergic Neurons/metabolism , Mice , Brain/physiology , Brain/cytology , Synapses/physiology , Synapses/metabolism , Axons/physiology , Axons/metabolism , Male
13.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38969499

ABSTRACT

The adult turtle spinal cord can generate multiple kinds of limb movements, including swimming, three forms of scratching, and limb withdrawal (flexion reflex), even without brain input and sensory feedback. There are many multifunctional spinal neurons, activated during multiple motor patterns, and some behaviorally specialized neurons, activated during only one. How do multifunctional and behaviorally specialized neurons each contribute to motor output? We analyzed in vivo intracellular recordings of multifunctional and specialized neurons. Neurons tended to spike in the same phase of the hip-flexor (HF) activity cycle during swimming and scratching, though one preferred opposite phases. During both swimming and scratching, a larger fraction of multifunctional neurons than specialized neurons were highly rhythmic. One group of multifunctional neurons was active during the HF-on phase and another during the HF-off phase. Thus, HF-extensor alternation may be generated by a subset of multifunctional spinal neurons during both swimming and scratching. Scratch-specialized neurons and flexion reflex-selective neurons may instead trigger their respective motor patterns, by biasing activity of multifunctional neurons. In phase-averaged membrane potentials of multifunctional neurons, trough phases were more highly correlated between swimming and scratching than peak phases, suggesting that rhythmic inhibition plays a greater role than rhythmic excitation. We also provide the first intracellular recording of a turtle swim-specialized neuron: tonically excited during swimming but inactive during scratching and flexion reflex. It displayed an excitatory postsynaptic potential following each swim-evoking electrical stimulus and thus may be an intermediary between reticulospinal axons and the swimming CPG they activate.


Subject(s)
Reflex , Spinal Cord , Swimming , Turtles , Animals , Turtles/physiology , Swimming/physiology , Spinal Cord/physiology , Reflex/physiology , Neurons/physiology , Action Potentials/physiology , Motor Neurons/physiology
14.
Front Cell Neurosci ; 18: 1421617, 2024.
Article in English | MEDLINE | ID: mdl-38994327

ABSTRACT

The basolateral amygdala plays pivotal roles in the regulation of fear and anxiety and these processes are profoundly modulated by different neuromodulatory systems that are recruited during emotional arousal. Recent studies suggest activities of BLA interneurons and inhibitory synaptic transmission in BLA principal cells are regulated by neuromodulators to influence the output and oscillatory network states of the BLA, and ultimately the behavioral expression of fear and anxiety. In this review, we first summarize a cellular mechanism of stress-induced anxiogenesis mediated by the interaction of glucocorticoid and endocannabinoid signaling at inhibitory synapses in the BLA. Then we discuss cell type-specific activity patterns induced by neuromodulators converging on the Gq signaling pathway in BLA perisomatic parvalbumin-expressing (PV) and cholecystokinin-expressing (CCK) basket cells and their effects on BLA network oscillations and fear learning.

15.
Eur J Neurosci ; 60(5): 4937-4953, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39080914

ABSTRACT

Cholinergic interneurons (ChIs) act as master regulators of striatal output, finely tuning neurotransmission to control motivated behaviours. ChIs are a cellular target of many peptide and hormonal neuromodulators, including corticotropin-releasing factor, opioids, insulin and leptin, which can influence an animal's behaviour by signalling stress, pleasure, pain and nutritional status. However, little is known about how sex hormones via estrogen receptors influence the function of these other neuromodulators. Here, we performed in situ hybridisation on mouse striatal tissue to characterise the effect of sex and sex hormones on choline acetyltransferase (Chat), estrogen receptor alpha (Esr1) and corticotropin-releasing factor type 1 receptor (Crhr1) expression. Although we did not detect sex differences in ChAT protein levels in the dorsal striatum or nucleus accumbens, we found that female mice have more Chat mRNA-expressing neurons than males in both the dorsal striatum and nucleus accumbens. At the population level, we observed a sexually dimorphic distribution of Esr1- and Crhr1-expressing ChIs in the ventral striatum that was negatively correlated in intact females, which was abolished by ovariectomy and not present in males. Only in the NAc did we find a significant population of ChIs that co-express Crhr1 and Esr1 in females and to a lesser extent in males. At the cellular level, Crhr1 and Esr1 transcript levels were negatively correlated only during the estrus phase in females, indicating that changes in sex hormone levels can modulate the interaction between Crhr1 and Esr1 mRNA levels.


Subject(s)
Cholinergic Neurons , Corticotropin-Releasing Hormone , Estrogen Receptor alpha , Estrogens , Interneurons , Nucleus Accumbens , Receptors, Corticotropin-Releasing Hormone , Animals , Male , Nucleus Accumbens/metabolism , Female , Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Interneurons/metabolism , Estrogen Receptor alpha/metabolism , Mice , Cholinergic Neurons/metabolism , Estrogens/metabolism , Sex Characteristics , Mice, Inbred C57BL , Choline O-Acetyltransferase/metabolism , Ovariectomy
16.
Cell Rep ; 43(8): 114519, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39018243

ABSTRACT

Diverse neuron classes in hippocampal CA1 have been identified through the heterogeneity of their cellular/molecular composition. How these classes relate to hippocampal function and the network dynamics that support cognition in primates remains unclear. Here, we report inhibitory functional cell groups in CA1 of freely moving macaques whose diverse response profiles to network states and each other suggest distinct and specific roles in the functional microcircuit of CA1. In addition, pyramidal cells that were grouped by their superficial or deep layer position differed in firing rate, burstiness, and sharp-wave ripple-associated firing. They also showed strata-specific spike-timing interactions with inhibitory cell groups, suggestive of segregated neural populations. Furthermore, ensemble recordings revealed that cell assemblies were preferentially organized according to these strata. These results suggest that hippocampal CA1 in freely moving macaques bears a sublayer-specific circuit organization that may shape its role in cognition.


Subject(s)
CA1 Region, Hippocampal , Pyramidal Cells , Animals , Pyramidal Cells/physiology , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/cytology , Action Potentials/physiology , Male , Nerve Net/physiology
17.
Biol Psychiatry ; 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38950809

ABSTRACT

BACKGROUND: Exaggerated responses to sensory stimuli, a hallmark of fragile X syndrome, contribute to anxiety and learning challenges. Sensory hypersensitivity is recapitulated in the Fmr1 knockout (KO) mouse model of fragile X syndrome. Recent studies in Fmr1 KO mice have demonstrated differences in the activity of cortical interneurons and a delayed switch in the polarity of GABA (gamma-aminobutyric acid) signaling during development. Previously, we reported that blocking the chloride transporter NKCC1 with the diuretic bumetanide could rescue synaptic circuit phenotypes in the primary somatosensory cortex (S1) of Fmr1 KO mice. However, it remains unknown whether bumetanide can rescue earlier circuit phenotypes or sensory hypersensitivity in Fmr1 KO mice. METHODS: We used acute and chronic systemic administration of bumetanide in Fmr1 KO mice and performed in vivo 2-photon calcium imaging to record neuronal activity, while tracking mouse behavior with high-resolution videos. RESULTS: We demonstrated that layer 2/3 pyramidal neurons in the S1 of Fmr1 KO mice showed a higher frequency of synchronous events on postnatal day 6 than wild-type controls. This was reversed by acute administration of bumetanide. Furthermore, chronic bumetanide treatment (postnatal days 5-14) restored S1 circuit differences in Fmr1 KO mice, including reduced neuronal adaptation to repetitive whisker stimulation, and ameliorated tactile defensiveness. Bumetanide treatment also rectified the reduced feedforward inhibition of layer 2/3 neurons in the S1 and boosted the circuit participation of parvalbumin interneurons. CONCLUSIONS: This further supports the notion that synaptic, circuit, and sensory behavioral phenotypes in Fmr1 KO can be mitigated by inhibitors of NKCC1, such as the Food and Drug Administration-approved diuretic bumetanide.

18.
Cell Rep ; 43(8): 114540, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39058595

ABSTRACT

Long-term synaptic plasticity at glutamatergic synapses on striatal spiny projection neurons (SPNs) is central to learning goal-directed behaviors and habits. Our studies reveal that SPNs manifest a heterosynaptic, nitric oxide (NO)-dependent form of long-term postsynaptic depression of glutamatergic SPN synapses (NO-LTD) that is preferentially engaged at quiescent synapses. Plasticity is gated by Ca2+ entry through CaV1.3 Ca2+ channels and phosphodiesterase 1 (PDE1) activation, which blunts intracellular cyclic guanosine monophosphate (cGMP) and NO signaling. Both experimental and simulation studies suggest that this Ca2+-dependent regulation of PDE1 activity allows for local regulation of dendritic cGMP signaling. In a mouse model of Parkinson disease (PD), NO-LTD is absent because of impaired interneuronal NO release; re-balancing intrastriatal neuromodulatory signaling restores NO release and NO-LTD. Taken together, these studies provide important insights into the mechanisms governing NO-LTD in SPNs and its role in psychomotor disorders such as PD.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1 , Neuronal Plasticity , Neurons , Synapses , Animals , Synapses/metabolism , Neuronal Plasticity/physiology , Mice , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Corpus Striatum/metabolism , Cyclic GMP/metabolism , Glutamic Acid/metabolism , Calcium/metabolism , Mice, Inbred C57BL , Male , Long-Term Synaptic Depression/physiology
19.
eNeuro ; 11(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39084907

ABSTRACT

The integration of spatial information in the mammalian dentate gyrus (DG) is critical to navigation. Indeed, DG granule cells (DGCs) rely upon finely balanced inhibitory neurotransmission in order to respond appropriately to specific spatial inputs. This inhibition arises from a heterogeneous population of local GABAergic interneurons (INs) that activate both fast, ionotropic GABAA receptors (GABAAR) and slow, metabotropic GABAB receptors (GABABR), respectively. GABABRs in turn inhibit pre- and postsynaptic neuronal compartments via temporally long-lasting G-protein-dependent mechanisms. The relative contribution of each IN subtype to network level GABABR signal setting remains unknown. However, within the DG, the somatostatin (SSt) expressing IN subtype is considered crucial in coordinating appropriate feedback inhibition on to DGCs. Therefore, we virally delivered channelrhodopsin 2 to the DG in order to obtain control of this specific SSt IN subpopulation in male and female adult mice. Using a combination of optogenetic activation and pharmacology, we show that SSt INs strongly recruit postsynaptic GABABRs to drive greater inhibition in DGCs than GABAARs at physiological membrane potentials. Furthermore, we show that in the adult mouse DG, postsynaptic GABABR signaling is predominantly regulated by neuronal GABA uptake and less so by astrocytic mechanisms. Finally, we confirm that activation of SSt INs can also recruit presynaptic GABABRs, as has been shown in neocortical circuits. Together, these data reveal that GABABR signaling allows SSt INs to control DG activity and may constitute a key mechanism for gating spatial information flow within hippocampal circuits.


Subject(s)
Dentate Gyrus , Interneurons , Receptors, GABA-B , Somatostatin , Animals , Somatostatin/metabolism , Interneurons/metabolism , Interneurons/physiology , Dentate Gyrus/metabolism , Receptors, GABA-B/metabolism , Male , Female , Optogenetics , Mice, Inbred C57BL , Mice , Mice, Transgenic , gamma-Aminobutyric Acid/metabolism , Synapses/metabolism
20.
bioRxiv ; 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39005261

ABSTRACT

The CA3 region is central to hippocampal function during learning and memory and has a unique connectivity. CA3 pyramidal neurons are the targets of huge, excitatory mossy fiber synapses from DG axons and have a high degree of excitatory recurrent connectivity. Thus, inhibition likely plays an outsized importance in constraining excitation and shaping CA3 ensembles during learning and memory. Here, we investigate the function of a never-before studied set of dendrite-targeting, GABAergic neurons defined by expression of the synaptic adhesion molecule, Kirrel3. We discovered that activating Kirrel3-expressing GABAergic neurons specifically impairs memory discrimination and inhibits CA3 pyramidal neurons in novel contexts. Kirrel3 is required for DG-to-GABA synapse formation and variants in Kirrel3 are strong risk factors for neurodevelopmental disorders. Thus, our work suggests that Kirrel3-GABA neurons are a critical source of feed-forward inhibition from DG to CA3 during the encoding and retrieval of contextual memories, a function which may be specifically disrupted in some brain disorders.

SELECTION OF CITATIONS
SEARCH DETAIL