Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Neurosci ; 42(24): 4828-4840, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35534225

ABSTRACT

The functions of cortical networks are progressively established during development by series of events shaping the neuronal connectivity. Synaptic elimination, which consists of removing the supernumerary connections generated during the earlier stages of cortical development, is one of the latest stages in neuronal network maturation. The semaphorin 3F coreceptors neuropilin 2 (Nrp2) and plexin-A3 (PlxnA3) may play an important role in the functional maturation of the cerebral cortex by regulating the excess dendritic spines on cortical excitatory neurons. Yet, the identity of the connections eliminated under the control of Nrp2/PlxnA3 signaling is debated, and the importance of this synaptic refinement for cortical functions remains poorly understood. Here, we show that Nrp2/PlxnA3 controls the spine densities in layer 4 (L4) and on the apical dendrite of L5 neurons of the sensory and motor cortices. Using a combination of neuroanatomical, ex vivo electrophysiology, and in vivo functional imaging techniques in Nrp2 and PlxnA3 KO mice of both sexes, we disprove the hypothesis that Nrp2/PlxnA3 signaling is required to maintain the ectopic thalamocortical connections observed during embryonic development. We also show that the absence of Nrp2/PlxnA3 signaling leads to the hyperexcitability and excessive synchronization of the neuronal activity in L5 and L4 neuronal networks, suggesting that this system could participate in the refinement of the recurrent corticocortical connectivity in those layers. Altogether, our results argue for a role of semaphorin-Nrp2/PlxnA3 signaling in the proper maturation and functional connectivity of the cerebral cortex, likely by controlling the refinement of recurrent corticocortical connections.SIGNIFICANCE STATEMENT The function of a neuronal circuit is mainly determined by the connections that neurons establish with one another during development. Understanding the mechanisms underlying the establishment of the functional connectivity is fundamental to comprehend how network functions are implemented, and to design treatments aiming at restoring damaged neuronal circuits. Here, we show that the cell surface receptors for the family of semaphorin guidance cues neuropilin 2 (Nrp2) and plexin-A3 (PlxnA3) play an important role in shaping the functional connectivity of the cerebral cortex likely by trimming the recurrent connections in layers 4 and 5. By removing the supernumerary inputs generated during early development, Nrp2/PlxnA3 signaling reduces the neuronal excitability and participates in the maturation of the cortical network functions.


Subject(s)
Neuropilin-2 , Semaphorins , Animals , Cell Adhesion Molecules , Cerebral Cortex/metabolism , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins , Neuropilin-2/metabolism , Semaphorins/metabolism
2.
Nature ; 542(7641): 362-366, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28178232

ABSTRACT

Malignant neoplasms evolve in response to changes in oncogenic signalling. Cancer cell plasticity in response to evolutionary pressures is fundamental to tumour progression and the development of therapeutic resistance. Here we determine the molecular and cellular mechanisms of cancer cell plasticity in a conditional oncogenic Kras mouse model of pancreatic ductal adenocarcinoma (PDAC), a malignancy that displays considerable phenotypic diversity and morphological heterogeneity. In this model, stochastic extinction of oncogenic Kras signalling and emergence of Kras-independent escaper populations (cells that acquire oncogenic properties) are associated with de-differentiation and aggressive biological behaviour. Transcriptomic and functional analyses of Kras-independent escapers reveal the presence of Smarcb1-Myc-network-driven mesenchymal reprogramming and independence from MAPK signalling. A somatic mosaic model of PDAC, which allows time-restricted perturbation of cell fate, shows that depletion of Smarcb1 activates the Myc network, driving an anabolic switch that increases protein metabolism and adaptive activation of endoplasmic-reticulum-stress-induced survival pathways. Increased protein turnover renders mesenchymal sub-populations highly susceptible to pharmacological and genetic perturbation of the cellular proteostatic machinery and the IRE1-α-MKK4 arm of the endoplasmic-reticulum-stress-response pathway. Specifically, combination regimens that impair the unfolded protein responses block the emergence of aggressive mesenchymal subpopulations in mouse and patient-derived PDAC models. These molecular and biological insights inform a potential therapeutic strategy for targeting aggressive mesenchymal features of PDAC.


Subject(s)
Mesoderm/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Endoplasmic Reticulum Stress/genetics , Female , Genes, myc , Genes, ras , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Male , Mesoderm/metabolism , Mice , Mosaicism , Oncogene Protein p55(v-myc)/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SMARCB1 Protein/deficiency , SMARCB1 Protein/metabolism , Transcriptome/genetics , Gemcitabine
3.
J Neurosci ; 39(24): 4727-4737, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30952811

ABSTRACT

The main excitatory inputs to the striatum arising from the cortex and the thalamus innervate both striatal spiny projection neurons and interneurons. These glutamatergic inputs to striatal GABAergic interneurons have been suggested to regulate the spike timing of striatal projection neurons via feedforward inhibition. Understanding how different excitatory inputs are integrated within the striatal circuitry and how they regulate striatal output is crucial for understanding basal ganglia function and related behaviors. Here, using VGLUT2 mice from both sexes, we report the existence of a glutamatergic projection from the mesencephalic locomotor region to the striatum that avoids the spiny neurons and selectively innervates interneurons. Specifically, optogenetic activation of glutamatergic axons from the pedunculopontine nucleus induced monosynaptic excitation in most recorded striatal cholinergic interneurons and GABAergic fast-spiking interneurons. Optogenetic stimulation in awake head-fixed mice consistently induced an increase in the firing rate of putative cholinergic interneurons and fast-spiking interneurons. In contrast, this stimulation did not induce excitatory responses in spiny neurons but rather disynaptic inhibitory responses ex vivo and a decrease in their firing rate in vivo, suggesting a feedforward mechanism mediating the inhibition of spiny projection neurons through the selective activation of striatal interneurons. Furthermore, unilateral stimulation of pedunculopontine nucleus glutamatergic axons in the striatum induced ipsilateral head rotations consistent with the inhibition of striatal output neurons. Our results demonstrate the existence of a unique interneuron-specific midbrain glutamatergic input to the striatum that exclusively recruits feedforward inhibition mechanisms.SIGNIFICANCE STATEMENT Glutamatergic inputs to the striatum have been shown to target both striatal projection neurons and interneurons and have been proposed to regulate spike timing of the projection neurons in part through feedforward inhibition. Here, we reveal the existence of a midbrain source of glutamatergic innervation to the striatum, originating in the pedunculopontine nucleus. Remarkably, this novel input selectively targets striatal interneurons, avoiding the projection neurons. Furthermore, we show that this selective innervation of interneurons can regulate the firing of the spiny projection neurons and inhibit the striatal output via feedforward inhibition. Together, our results describe a unique source of excitatory innervation to the striatum which selectively recruits feedforward inhibition of spiny neurons without any accompanying excitation.


Subject(s)
Interneurons/physiology , Neostriatum/cytology , Neostriatum/physiology , Neural Inhibition/physiology , Neurons/physiology , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/physiology , gamma-Aminobutyric Acid/physiology , Animals , Animals, Genetically Modified , Axons/physiology , Basal Ganglia/physiology , Female , Locomotion/physiology , Male , Mesencephalon/physiology , Mice , Nerve Net/cytology , Nerve Net/physiology , Optogenetics , Parasympathetic Nervous System/physiology , Vesicular Glutamate Transport Protein 2/genetics
4.
J Neurosci ; 39(45): 8845-8859, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31541021

ABSTRACT

The striatum represents the main input structure of the basal ganglia, receiving massive excitatory input from the cortex and the thalamus. The development and maintenance of cortical input to the striatum is crucial for all striatal function including many forms of sensorimotor integration, learning, and action control. The molecular mechanisms regulating the development and maintenance of corticostriatal synaptic transmission are unclear. Here we show that the guidance cue, Semaphorin 3F and its receptor Neuropilin 2 (Nrp2), influence dendritic spine maintenance, corticostriatal short-term plasticity, and learning in adult male and female mice. We found that Nrp2 is enriched in adult layer V pyramidal neurons, corticostriatal terminals, and in developing and adult striatal spiny projection neurons (SPNs). Loss of Nrp2 increases SPN excitability and spine number, reduces short-term facilitation at corticostriatal synapses, and impairs goal-directed learning in an instrumental task. Acute deletion of Nrp2 selectively in adult layer V cortical neurons produces a similar increase in the number of dendritic spines and presynaptic modifications at the corticostriatal synapse in the Nrp2-/- mouse, but does not affect the intrinsic excitability of SPNs. Furthermore, conditional loss of Nrp2 impairs sensorimotor learning on the accelerating rotarod without affecting goal-directed instrumental learning. Collectively, our results identify Nrp2 signaling as essential for the development and maintenance of the corticostriatal pathway and may shed novel insights on neurodevelopmental disorders linked to the corticostriatal pathway and Semaphorin signaling.SIGNIFICANCE STATEMENT The corticostriatal pathway controls sensorimotor, learning, and action control behaviors and its dysregulation is linked to neurodevelopmental disorders, such as autism spectrum disorder (ASD). Here we demonstrate that Neuropilin 2 (Nrp2), a receptor for the axon guidance cue semaphorin 3F, has important and previously unappreciated functions in the development and adult maintenance of dendritic spines on striatal spiny projection neurons (SPNs), corticostriatal short-term plasticity, intrinsic physiological properties of SPNs, and learning in mice. Our findings, coupled with the association of Nrp2 with ASD in human populations, suggest that Nrp2 may play an important role in ASD pathophysiology. Overall, our work demonstrates Nrp2 to be a key regulator of corticostriatal development, maintenance, and function, and may lead to better understanding of neurodevelopmental disease mechanisms.


Subject(s)
Cerebral Cortex/metabolism , Conditioning, Operant , Corpus Striatum/metabolism , Neuropilin-2/metabolism , Synaptic Transmission , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Corpus Striatum/growth & development , Corpus Striatum/physiology , Dendritic Spines/metabolism , Dendritic Spines/physiology , Female , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurogenesis , Neuropilin-2/genetics , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology
5.
J Neurosci ; 38(25): 5688-5699, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29789374

ABSTRACT

The recent availability of different transgenic mouse lines coupled with other modern molecular techniques has led to the discovery of an unexpectedly large cellular diversity and synaptic specificity in striatal interneuronal circuitry. Prior research has described three spontaneously active interneuron types in mouse striatal slices: the cholinergic interneuron, the neuropeptide Y-low threshold spike interneuron, and the tyrosine hydroxylase interneurons (THINs). Using transgenic Htr3a-Cre mice, we now characterize a fourth population of spontaneously active striatal GABAergic interneurons termed spontaneously active bursty interneurons (SABIs) because of their unique burst-firing pattern in cell-attached recordings. Although they bear some qualitative similarity in intrinsic electrophysiological properties to THINs in whole-cell recordings, detailed analysis revealed significant differences in many intrinsic properties and in their morphology. Furthermore, all previously identified striatal GABAergic interneurons have been shown to innervate striatal spiny projection neurons (SPNs), contributing to the suggestion that the principal function of striatal GABAergic interneurons is to provide feedforward inhibition to SPNs. Here, very surprisingly, paired recordings show that SABIs do not innervate SPNs significantly. Further, optogenetic inhibition of striatal Htr3a-Cre interneurons triggers barrages of IPSCs in SPNs. We hypothesize that these IPSCs result from disinhibition of a population of GABAergic interneurons with activity that is constitutively suppressed by the SABIs. We suggest that the SABIs represent the first example of a striatal interneuron-selective interneuron and, further, that their existence, along with previously defined interneuronal networks, may participate in the formation of SPN ensembles observed by others.SIGNIFICANCE STATEMENT Before ∼2010, the main function of the three known subtypes of striatal GABAergic interneurons was assumed to mediate feedforward inhibition of the spiny neurons (SPNs). During the past decade, we and others have described several novel populations of striatal GABAergic interneurons and their synaptic connections and have shown that striatal interneurons and SPNs interact through extensive and highly cell-type-specific connections that form specialized networks. Here, we describe a novel population of striatal GABAergic interneuron and provide several lines of evidence suggesting that it represents the first interneuron-selective interneuron in striatum. Striatal interneurons and their synaptic connections are suggested to play an important role in the formation of ensembles of striatal SPNs interconnected by inhibitory axon collaterals.


Subject(s)
GABAergic Neurons/cytology , Interneurons/cytology , Neostriatum/cytology , Animals , GABAergic Neurons/physiology , Interneurons/physiology , Mice
6.
Eur J Neurosci ; 49(5): 593-603, 2019 03.
Article in English | MEDLINE | ID: mdl-29480942

ABSTRACT

The striatum constitutes the main input structure of the basal ganglia and receives two major excitatory glutamatergic inputs, from the cortex and the thalamus. Excitatory cortico- and thalamostriatal connections innervate the principal neurons of the striatum, the spiny projection neurons (SPNs), which constitute the main cellular input as well as the only output of the striatum. In addition, corticostriatal and thalamostriatal inputs also innervate striatal interneurons. Some of these inputs have been very well studied, for example the thalamic innervation of cholinergic interneurons and the cortical innervation of striatal fast-spiking interneurons, but inputs to most other GABAergic interneurons remain largely unstudied, due in part to the relatively recent identification and characterization of many of these interneurons. In this review, we will discuss and reconcile some older as well as more recent data on the extrinsic excitatory inputs to striatal interneurons. We propose that the traditional feed-forward inhibitory model of the cortical input to the fast-spiking interneuron then inhibiting the SPN, often assumed to be the prototype of the main functional organization of striatal interneurons, is incomplete. We provide evidence that the extrinsic innervation of striatal interneurons is not uniform but shows great cell-type specificity. In addition, we will review data showing that striatal interneurons are themselves interconnected in a highly cell-type-specific manner. These data suggest that the impact of the extrinsic inputs on striatal activity critically depends on synaptic interactions within interneuronal circuitry.


Subject(s)
Cerebral Cortex/physiology , Cholinergic Neurons/physiology , Corpus Striatum/physiology , Electrophysiological Phenomena/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Nerve Net/physiology , Neurons, Afferent/physiology , Thalamus/physiology , Animals
7.
Eur J Neurosci ; 50(4): 2653-2662, 2019 08.
Article in English | MEDLINE | ID: mdl-30941837

ABSTRACT

The striatum mediates a broad range of cognitive and motor functions. Within the striatum, recently discovered tyrosine hydroxylase expressing interneurons (THINs) provide a source of intrastriatal synaptic connectivity that is critical for regulating striatal activity, yet the role of THIN's in behavior remains unknown. Given the important role of the striatum in reward-based behaviors, we investigated whether loss of striatal THINs would impact instrumental behavior in mice. We selectively ablated striatal THINs in TH-Cre mice using chemogenetic techniques, and then tested THIN-lesioned or control mice on three reward-based striatal-dependent instrumental tests: (a) progressive ratio test; (b) choice test following selective-satiety induced outcome devaluation; (c) outcome reinstatement test. Both striatal-THIN-lesioned and control mice acquired an instrumental response for flavored food pellets, and their behavior did not differ in the progressive ratio test, suggesting intact effort to obtain rewards. However, striatal THIN lesions markedly impaired choice performance following selective-satiety induced outcome devaluation. Unlike control mice, THIN-lesioned mice did not adjust their choice of actions following a change in outcome value. In the outcome reinstatement test THIN-lesioned and control mice showed response invigoration by outcome presentation, suggesting the incentive properties of outcomes were not disrupted by THIN lesions. Overall, we found that striatal THIN lesions selectively impaired goal-directed behavior, while preserving motoric and appetitive behaviors. These findings are the first to describe a function of striatal THINs in reward-based behavior, and further illustrate the important role for intrastriatal interneuronal connectivity in behavioral functions ascribed to the striatum more generally.


Subject(s)
Conditioning, Operant , Interneurons/pathology , Neostriatum/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Animals , Appetitive Behavior , Choice Behavior , Extinction, Psychological , Goals , Interneurons/enzymology , Male , Mice , Mice, Transgenic , Motor Activity , Neostriatum/cytology , Neostriatum/enzymology , Psychomotor Performance , Reinforcement Schedule , Reward
8.
J Neurosci Res ; 97(12): 1491-1502, 2019 12.
Article in English | MEDLINE | ID: mdl-31102306

ABSTRACT

The classical view of striatal GABAergic interneuron function has been that they operate as largely independent, parallel, feedforward inhibitory elements providing inhibitory inputs to spiny projection neurons (SPNs). Much recent evidence has shown that the extrinsic innervation of striatal interneurons is not indiscriminate but rather very specific, and that striatal interneurons are themselves interconnected in a cell type-specific manner. This suggests that the ultimate effect of extrinsic inputs on striatal neuronal activity depends critically on synaptic interactions within interneuronal circuitry. Here, we compared the cortical and thalamic input to two recently described subtypes of striatal GABAergic interneurons, tyrosine hydroxylase-expressing interneurons (THINs), and spontaneously active bursty interneurons (SABIs) using transgenic TH-Cre and Htr3a-Cre mice of both sexes. Our results show that both THINs and SABIs receive strong excitatory input from the motor cortex and the thalamic parafascicular nucleus. Cortical optogenetic stimulation also evokes disynaptic inhibitory GABAergic responses in THINs but not in SABIs. In contrast, optogenetic stimulation of the parafascicular nucleus induces disynaptic inhibitory responses in both interneuron populations. However, the short-term plasticity of these disynaptic inhibitory responses is different suggesting the involvement of different intrastriatal microcircuits. Altogether, our results point to highly specific interneuronal circuits that are selectively engaged by different excitatory inputs.


Subject(s)
Cerebral Cortex/physiology , Corpus Striatum/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Intralaminar Thalamic Nuclei/physiology , Membrane Potentials , Action Potentials , Animals , Excitatory Postsynaptic Potentials , Female , Inhibitory Postsynaptic Potentials , Male , Mice, Transgenic , Neural Pathways/physiology , Optogenetics
9.
J Neurosci ; 36(36): 9505-11, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27605623

ABSTRACT

UNLABELLED: Synchronous optogenetic activation of striatal cholinergic interneurons ex vivo produces a disynaptic inhibition of spiny projection neurons composed of biophysically distinct GABAAfast and GABAAslow components. This has been shown to be due, at least in part, to activation of nicotinic receptors on GABAergic NPY-neurogliaform interneurons that monosynaptically inhibit striatal spiny projection neurons. Recently, it has been proposed that a significant proportion of this inhibition is actually mediated by activation of presynaptic nicotinic receptors on nigrostriatal terminals that evoke GABA release from the terminals of the dopaminergic nigrostriatal pathway. To disambiguate these the two mechanisms, we crossed mice in which channelrhodopsin is endogenously expressed in cholinergic neurons with Htr3a-Cre mice, in which Cre is selectively targeted to several populations of striatal GABAergic interneurons, including the striatal NPY-neurogliaform interneuron. Htr3a-Cre mice were then virally transduced to express halorhodopsin to allow activation of channelrhodopsin and halorhodopsin, individually or simultaneously. Thus we were able to optogenetically disconnect the interneuron-spiny projection neuron (SPN) cell circuit on a trial-by-trial basis. As expected, optogenetic activation of cholinergic interneurons produced inhibitory currents in SPNs. During simultaneous inhibition of GABAergic interneurons with halorhodopsin, we observed a large, sometimes near complete reduction in both fast and slow components of the cholinergic-evoked inhibition, and a delay in IPSC latency. This demonstrates that the majority of cholinergic-evoked striatal GABAergic inhibition is derived from GABAergic interneurons. These results also reinforce the notion that a semiautonomous circuit of striatal GABAergic interneurons is responsible for transmitting behaviorally relevant cholinergic signals to spiny projection neurons. SIGNIFICANCE STATEMENT: The circuitry between neurons of the striatum has been recently described to be far more complex than originally imagined. One example of this phenomenon is that striatal cholinergic interneurons have been shown to provide intrinsic nicotinic excitation of local GABAergic interneurons, which then inhibit the projection neurons of the striatum. As deficits of cholinergic interneurons are reported in patients with Tourette syndrome, the normal functions of these interneurons are of great interest. Whether this novel route of nicotinic input constitutes a major output of cholinergic interneurons remains unknown. The study addressed this question using excitatory and inhibitory optogenetic technology, so that cholinergic interneurons could be selectively activated and GABAergic interneurons selectively inhibited to determine the causal relationship in this circuit.


Subject(s)
Cholinergic Neurons/physiology , Corpus Striatum/cytology , GABAergic Neurons/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Channelrhodopsins , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/pharmacology , Cholinergic Neurons/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABAergic Neurons/drug effects , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Nerve Growth Factor/metabolism , Nerve Net/drug effects , Neural Inhibition/drug effects , Neuropeptide Y/metabolism , Patch-Clamp Techniques , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Neurosci ; 35(16): 6584-99, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904808

ABSTRACT

Striatal GABAergic interneurons that express the gene for tyrosine hydroxylase (TH) have been identified previously by several methods. Although generally assumed to be dopaminergic, possibly serving as a compensatory source of dopamine (DA) in Parkinson's disease, this assumption has never been tested directly. In TH-Cre mice whose nigrostriatal pathway had been eliminated unilaterally with 6-hydroxydopamine, we injected a Cre-dependent virus coding for channelrhodopsin-2 and enhanced yellow fluorescent protein unilaterally into the unlesioned midbrain or bilaterally into the striatum. Fast-scan cyclic voltammetry in striatal slices revealed that both optical and electrical stimulation readily elicited DA release in control striata but not from contralateral striata when nigrostriatal neurons were transduced. In contrast, neither optical nor electrical stimulation could elicit striatal DA release in either the control or lesioned striata when the virus was injected directly into the striatum transducing only striatal TH interneurons. This demonstrates that striatal TH interneurons do not release DA. Fluorescence immunocytochemistry in enhanced green fluorescent protein (EGFP)-TH mice revealed colocalization of DA, l-amino acid decarboxylase, the DA transporter, and vesicular monoamine transporter-2 with EGFP in midbrain dopaminergic neurons but not in any of the striatal EGFP-TH interneurons. Optogenetic activation of striatal EGFP-TH interneurons produced strong GABAergic inhibition in all spiny neurons tested. These results indicate that striatal TH interneurons are not dopaminergic but rather are a type of GABAergic interneuron that expresses TH but none of the other enzymes or transporters necessary to operate as dopaminergic neurons and exert widespread GABAergic inhibition onto direct and indirect spiny neurons.


Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Aromatic-L-Amino-Acid Decarboxylases/metabolism , Corpus Striatum/physiology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Electric Stimulation , Female , GABAergic Neurons/physiology , Interneurons/physiology , Male , Mesencephalon/metabolism , Mice , Mice, Transgenic , Neural Inhibition/physiology , Optogenetics , Photic Stimulation , Vesicular Monoamine Transport Proteins/metabolism
11.
Eur J Neurosci ; 42(2): 1764-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25865337

ABSTRACT

Previous work suggests that neostriatal cholinergic interneurons control the activity of several classes of GABAergic interneurons through fast nicotinic receptor-mediated synaptic inputs. Although indirect evidence has suggested the existence of several classes of interneurons controlled by this mechanism, only one such cell type, the neuropeptide-Y-expressing neurogliaform neuron, has been identified to date. Here we tested the hypothesis that in addition to the neurogliaform neurons that elicit slow GABAergic inhibitory responses, another interneuron type exists in the striatum that receives strong nicotinic cholinergic input and elicits conventional fast GABAergic synaptic responses in projection neurons. We obtained in vitro slice recordings from double transgenic mice in which Channelrhodopsin-2 was natively expressed in cholinergic neurons and a population of serotonin receptor-3a-Cre-expressing GABAergic interneurons were visualized with tdTomato. We show that among the targeted GABAergic interneurons a novel type of interneuron, termed the fast-adapting interneuron, can be identified that is distinct from previously known interneurons based on immunocytochemical and electrophysiological criteria. We show using optogenetic activation of cholinergic inputs that fast-adapting interneurons receive a powerful supra-threshold nicotinic cholinergic input in vitro. Moreover, fast adapting neurons are densely connected to projection neurons and elicit fast, GABAA receptor-mediated inhibitory postsynaptic current responses. The nicotinic receptor-mediated activation of fast-adapting interneurons may constitute an important mechanism through which cholinergic interneurons control the activity of projection neurons and perhaps the plasticity of their synaptic inputs when animals encounter reinforcing or otherwise salient stimuli.


Subject(s)
Adaptation, Physiological/physiology , Cholinergic Agents/pharmacology , Corpus Striatum/cytology , Fasting/physiology , GABAergic Neurons/physiology , Neurons/drug effects , Synaptic Potentials/physiology , Animals , Bacterial Proteins/genetics , Channelrhodopsins , Choline O-Acetyltransferase/metabolism , Dihydro-beta-Erythroidine/pharmacology , GABAergic Neurons/drug effects , Humans , In Vitro Techniques , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Mutation/genetics , Nerve Growth Factor/pharmacology , Patch-Clamp Techniques , Striatonigral Degeneration , Synaptic Potentials/drug effects
12.
J Neurosci ; 31(46): 16757-69, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090502

ABSTRACT

We investigated the properties of neostriatal neuropeptide Y (NPY)-expressing interneurons in transgenic GFP (green fluorescent protein)-NPY reporter mice. In vitro whole-cell recordings and biocytin staining demonstrated the existence of a novel class of neostriatal NPY-expressing GABAergic interneurons that exhibit electrophysiological, neurochemical, and morphological properties strikingly different from those of previously described NPY-containing, plateau-depolarization low-threshold spike (NPY-PLTS) interneurons. The novel NPY interneuron type (NPY-neurogliaform) differed from previously described NPY-PLTS interneurons by exhibiting a significantly lower input resistance and hyperpolarized membrane potential, regular, nonaccommodating spiking in response to depolarizing current injections, and an absence of plateau depolarizations or low-threshold spikes. NPY-neurogliaform interneurons were also easily distinguished morphologically by their dense, compact, and highly branched dendritic and local axonal arborizations that contrasted sharply with the sparse and extended axonal and dendritic arborizations of NPY-PLTS interneurons. Furthermore, NPY-neurogliaform interneurons did not express immunofluorescence for somatostatin or nitric oxide synthase that was ubiquitous in NPY-PLTS interneurons. IPSP/Cs could only rarely be elicited in spiny projection neurons (SPNs) in paired recordings with NPY-PLTS interneurons. In contrast, the probability of SPN innervation by NPY-neurogliaform interneurons was extremely high, the synapse very reliable (no failures were observed), and the resulting postsynaptic response was a slow, GABA(A) receptor-mediated IPSC that has not been previously described in striatum but that has been elicited from NPY-GABAergic neurogliaform interneurons in cortex and hippocampus. These properties suggest unique and distinctive roles for NPY-PLTS and NPY-neurogliaform interneurons in the integrative properties of the neostriatum.


Subject(s)
Corpus Striatum/cytology , Interneurons/classification , Interneurons/physiology , Neuropeptide Y/metabolism , Animals , Bicuculline/pharmacology , Cell Count , Cerebral Cortex/physiology , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Antagonists/pharmacology , Green Fluorescent Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factor/metabolism , Neural Pathways/physiology , Nitric Oxide Synthase/metabolism , Patch-Clamp Techniques , Quinoxalines/pharmacology , Somatostatin/metabolism
13.
Cell Rep ; 41(4): 111531, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288709

ABSTRACT

Cholinergic interneurons (CINs) are essential elements of striatal circuits and functions. Although acetylcholine signaling via muscarinic receptors (mAChRs) has been well studied, more recent data indicate that postsynaptic nicotinic receptors (nAChRs) located on striatal GABAergic interneurons (GINs) are equally critical. One example is that CIN stimulation induces large disynaptic inhibition of striatal projection neurons (SPNs) mediated by nAChR activation of GINs. Although these circuits are ideally positioned to modulate striatal output, the neurons involved are not definitively identified because of an incomplete mapping of CINs-GINs interconnections. Here, we show that CINs modulate four GINs populations via an intricate mechanism involving co-activation of presynaptic and postsynaptic mAChRs and nAChRs. Using optogenetics, we demonstrate the participation of tyrosine hydroxylase-expressing GINs in the disynaptic inhibition of SPNs via heterotypic electrical coupling with neurogliaform interneurons. Altogether, our results highlight the importance of CINs in regulating GINs microcircuits via complex synaptic/heterosynaptic mechanisms.


Subject(s)
Acetylcholine , Receptors, Nicotinic , Tyrosine 3-Monooxygenase , Corpus Striatum/physiology , Interneurons/physiology , Cholinergic Agents/pharmacology , Receptors, Muscarinic , Cholinergic Neurons/physiology
14.
J Neurosci ; 30(20): 6999-7016, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484642

ABSTRACT

Whole-cell recordings were obtained from tyrosine hydroxylase-expressing (TH(+)) neurons in striatal slices from bacterial artificial chromosome transgenic mice that synthesize enhanced green fluorescent protein (EGFP) selectively in neurons expressing TH transcriptional regulatory sequences. Stereological cell counting indicated that there were approximately 2700 EGFP-TH(+) neurons/striatum. Whole-cell recordings in striatal slices demonstrated that EGFP-TH(+) neurons comprise four electrophysiologically distinct neuron types whose electrophysiological properties have not been reported previously in striatum. EGFP-TH(+) neurons were identified in retrograde tracing studies as interneurons. Recordings from synaptically connected pairs of EGFP-TH(+) interneurons and spiny neurons showed that the interneurons elicited GABAergic IPSPs/IPSCs in spiny neurons powerful enough to significantly delay evoked spiking. EGFP-TH(+) interneurons responded to local or cortical stimulation with glutamatergic EPSPs. Local stimulation also elicited GABA(A) IPSPs, at least some of which arose from identified spiny neurons. Single-cell reverse transcription-PCR showed expression of VMAT1 in EGFP-TH(+) interneurons, consistent with previous suggestions that these interneurons may be dopaminergic as well as GABAergic. All four classes of interneurons were medium sized with modestly branching, varicose dendrites, and dense, highly varicose axon collateral fields. These data show for the first time that there exists in the normal rodent striatum a substantial population of TH(+)/GABAergic interneurons comprising four electrophysiologically distinct subtypes whose electrophysiological properties differ significantly from those of previously described striatal GABAergic interneurons. These interneurons are likely to play an important role in striatal function through fast GABAergic synaptic transmission in addition to, and independent of, their potential role in compensation for dopamine loss in experimental or idiopathic Parkinson's disease.


Subject(s)
Corpus Striatum/cytology , Neurons/cytology , Neurons/physiology , Synapses/physiology , Tyrosine 3-Monooxygenase/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Bicuculline/pharmacology , Calcium Channel Blockers/pharmacology , Cardiovascular Agents/pharmacology , Cell Count/methods , Colchicine/pharmacology , Corpus Striatum/drug effects , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Flufenamic Acid/pharmacology , GABA Antagonists/pharmacology , Green Fluorescent Proteins/genetics , In Vitro Techniques , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Mice, Transgenic , Neural Pathways/physiology , Neurons/classification , Nimodipine/pharmacology , Patch-Clamp Techniques/methods , Pyrimidines/pharmacology , Synaptic Transmission/drug effects , Time Factors , Tubulin Modulators/pharmacology , Tyrosine 3-Monooxygenase/genetics , Vesicular Monoamine Transport Proteins/genetics , Vesicular Monoamine Transport Proteins/metabolism
15.
J Neurosci ; 30(20): 7105-10, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484653

ABSTRACT

Recent evidence suggests the intriguing possibility that midbrain dopaminergic (DAergic) neurons may use fast glutamatergic transmission to communicate with their postsynaptic targets. Because of technical limitations, direct demonstration of the existence of this signaling mechanism has been limited to experiments using cell culture preparations that often alter neuronal function including neurotransmitter phenotype. Consequently, it remains uncertain whether glutamatergic signaling between DAergic neurons and their postsynaptic targets exists under physiological conditions. Here, using an optogenetic approach, we provide the first conclusive demonstration that mesolimbic DAergic neurons in mice release glutamate and elicit excitatory postsynaptic responses in projection neurons of the nucleus accumbens. In addition, we describe the properties of the postsynaptic glutamatergic responses of these neurons during experimentally evoked burst firing of DAergic axons that reproduce the reward-related phasic population activity of the mesolimbic projection. These observations indicate that, in addition to DAergic mechanisms, mesolimbic reward signaling may involve glutamatergic transmission.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Nucleus Accumbens/cytology , Signal Transduction/physiology , Animals , Dopamine Agents/pharmacology , Electric Stimulation/methods , Electrochemistry/methods , Excitatory Postsynaptic Potentials/drug effects , Female , Gene Transfer Techniques , In Vitro Techniques , Luminescent Proteins/genetics , Male , Mice , Neurons/drug effects , Patch-Clamp Techniques/methods , Photic Stimulation/methods , Signal Transduction/drug effects , Sodium Channel Blockers/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology
16.
J Neurosci ; 29(28): 8977-90, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605635

ABSTRACT

Recent studies have demonstrated that GABAergic synaptic transmission among neostriatal spiny projection neurons (SPNs) is strongly modulated by dopamine with individual connections exhibiting either D(1) receptor (D(1)R)-mediated facilitation or D(2) receptor (D(2)R)-mediated inhibition and, at least in some preparations, a subset of connections exhibiting both of these effects. In light of the cell type-specific expression of D(1a)R in striatonigral and D(2)R in striatopallidal neurons and the differential expression of the other D(1) and D(2) family dopamine receptors, we hypothesize that the nature of the dopaminergic modulation is specific to the types of SPNs that participate in the connection. Here the biophysical properties and dopaminergic modulation of intrastriatal connections formed by striatopallidal neurons were examined. Contrary to previous expectation, synapses formed by striatopallidal neurons were biophysically and pharmacologically heterogeneous. Two distinct types of axon collateral connections could be distinguished among striatopallidal neurons. The more common, small-amplitude connections (80%) exhibited mean IPSC amplitudes several times smaller than their less frequent large-amplitude counterparts, principally because of a smaller number of release sites involved. The two types of connections were also differentially regulated by dopamine. Small-amplitude connections exhibited strong and exclusively D(2)R-mediated presynaptic inhibition, whereas large-amplitude connections were unresponsive to dopamine. Synaptic connections from striatopallidal to striatonigral neurons exhibited exclusively D(2)R-mediated presynaptic inhibition that was similar to the regulation of small-amplitude connections between pairs of striatopallidal cells. Together, these findings demonstrate a previously unrecognized complexity in the organization and dopaminergic control of synaptic communication among SPNs.


Subject(s)
Axons/physiology , Dopamine/metabolism , Globus Pallidus/cytology , Neostriatum/cytology , Neurons/cytology , Synapses/physiology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Biophysics , Dopamine/pharmacology , Dopamine Agents/pharmacology , Electric Stimulation/methods , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/physiology , Neurons/classification , Patch-Clamp Techniques/methods , Receptors, Dopamine D2/genetics , Statistics, Nonparametric
17.
J Neurosci ; 28(41): 10386-98, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18842898

ABSTRACT

Most in vivo electrophysiological studies of substantia nigra have used rats. With the recent proliferation of the use of mice for in vitro neurophysiological studies because of the availability of various genetically modified strains to identify the roles of various channels and proteins in neuronal function, it is crucial to obtain data on in vivo responses in mice to verify that the in vitro results reflect functioning of systems comparable with those that have been well studied in rat. Inhibitory responses of rat nigral dopaminergic neurons by stimulation of afferents from striatum, globus pallidus, or pars reticulata have been shown to be mediated predominantly or exclusively by GABA(A) receptors. This is puzzling given the substantial expression of GABA(B) receptors and the ubiquitous appearance of GABA(B) synaptic responses in rat dopaminergic neurons in vitro. In the present study, we studied electrically evoked GABAergic inhibition in nigral dopaminergic neurons in C57BL/6J mice. Stimulation of the three major GABAergic inputs elicited stronger and longer-lasting inhibitory responses than those seen in rats. The early inhibition was GABA(A) mediated, whereas the later component, absent in rats, was GABA(B) mediated and selectively enhanced by GABA uptake inhibition. Striatal-evoked inhibition exhibited a slower onset and a weaker initial component compared with inhibition from globus pallidus or substantia nigra pars reticulata. These results are discussed with respect to differences in the size and neuronal density of the rat and mouse brain and the different sites of synaptic contact of the synapses from the three GABAergic afferents.


Subject(s)
Dopamine/metabolism , Neurons, Afferent/physiology , Neurons/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Substantia Nigra/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Corpus Striatum/cytology , Electric Stimulation/methods , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , GABA-B Receptor Antagonists , Globus Pallidus/cytology , Male , Mice , Mice, Inbred C57BL , Neural Inhibition , Neurons, Afferent/metabolism , Presynaptic Terminals/physiology , Rats , Substantia Nigra/cytology , Thalamus/physiology
18.
J Neural Transm Suppl ; (73): 71-90, 2009.
Article in English | MEDLINE | ID: mdl-20411769

ABSTRACT

Although substantia nigra dopaminergic neurons are spontaneously active both in vivo and in vitro, this activity does not depend on afferent input as these neurons express an endogenous calcium-dependent oscillatory mechanism sufficient to drive action potential generation. However, afferents to these neurons, a large proportion of them GABAergic and arising from other nuclei in the basal ganglia, play a crucial role in modulating the activity of dopaminergic neurons. In the absence of afferent activity or when in brain slices, dopaminergic neurons fire in a very regular, pacemaker-like mode. Phasic activity in GABAergic, glutamatergic, and cholinergic inputs modulates the pacemaker activity into two other modes. The most common is a random firing pattern in which interspike intervals assume a Poisson-like distribution, and a less common pattern, often in response to a conditioned stimulus or a reward in which the neurons fire bursts of 2-8 spikes time-locked to the stimulus. Typically in vivo, all three firing patterns are observed, intermixed, in single nigrostriatal neurons varying over time. Although the precise mechanism(s) underlying the burst are currently the focus of intensive study, it is obvious that bursting must be triggered by afferent inputs. Most of the afferents to substantia nigra pars compacta dopaminergic neurons comprise monosynaptic inputs from GABAergic projection neurons in the ipsilateral neostriatum, the globus pallidus, and the substantia nigra pars reticulata. A smaller fraction of the basal ganglia inputs, something less than 30%, are glutamatergic and arise principally from the ipsilateral subthalamic nucleus and pedunculopontine nucleus. The pedunculopontine nucleus also sends a cholinergic input to nigral dopaminergic neurons. The GABAergic pars reticulata projection neurons also receive inputs from all of these sources, in some cases relaying them disynaptically to the dopaminergic neurons, thereby playing a particularly significant role in setting and/or modulating the firing pattern of the nigrostriatal neurons.


Subject(s)
Basal Ganglia/physiology , Dopamine/metabolism , Neurons/physiology , Substantia Nigra/cytology , Animals , Humans , Models, Neurological , Nerve Net/physiology , Neural Pathways/physiology , Neurons/ultrastructure , Parkinson Disease/pathology , Synapses/physiology , Synapses/ultrastructure , gamma-Aminobutyric Acid/metabolism
19.
Brain Res Rev ; 58(2): 272-81, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18054796

ABSTRACT

There are two distinct inhibitory GABAergic circuits in the neostriatum. The feedforward circuit consists of a relatively small population of GABAergic interneurons that receives excitatory input from the neocortex and exerts monosynaptic inhibition onto striatal spiny projection neurons. The feedback circuit comprises the numerous spiny projection neurons and their interconnections via local axon collaterals. This network has long been assumed to provide the majority of striatal GABAergic inhibition and to sharpen and shape striatal output through lateral inhibition, producing increased activity in the most strongly excited spiny cells at the expense of their less strongly excited neighbors. Recent results, mostly from recording experiments of synaptically connected pairs of neurons, have revealed that the two GABAergic circuits differ markedly in terms of the total number of synapses made by each, the strength of the postsynaptic response detected at the soma, the extent of presynaptic convergence and divergence and the net effect of the activation of each circuit on the postsynaptic activity of the spiny neuron. These data have revealed that the feedforward inhibition is powerful and widespread, with spiking in a single interneuron being capable of significantly delaying or even blocking the generation of spikes in a large number of postsynaptic spiny neurons. In contrast, the postsynaptic effects of spiking in a single presynaptic spiny neuron on postsynaptic spiny neurons are weak when measured at the soma, and unable to significantly affect spike timing or generation. Further, reciprocity of synaptic connections between spiny neurons is only rarely observed. These results suggest that the bulk of the fast inhibition that has the strongest effects on spiny neuron spike timing comes from the feedforward interneuronal system whereas the axon collateral feedback system acts principally at the dendrites to control local excitability as well as the overall level of activity of the spiny neuron.


Subject(s)
Feedback/physiology , Neostriatum/cytology , Neural Inhibition/physiology , Neurons/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Neural Pathways/physiology , Neurons/classification
20.
Curr Biol ; 29(8): 1313-1323.e5, 2019 04 22.
Article in English | MEDLINE | ID: mdl-30982651

ABSTRACT

The striatum is the main input nucleus of the basal ganglia and is a key site of sensorimotor integration. While the striatum receives extensive excitatory afferents from the cerebral cortex, the influence of different cortical areas on striatal circuitry and behavior is unknown. Here, we find that corticostriatal inputs from whisker-related primary somatosensory (S1) and motor (M1) cortex differentially innervate projection neurons and interneurons in the dorsal striatum and exert opposing effects on sensory-guided behavior. Optogenetic stimulation of S1-corticostriatal afferents in ex vivo recordings produced larger postsynaptic potentials in striatal parvalbumin (PV)-expressing interneurons than D1- or D2-expressing spiny projection neurons (SPNs), an effect not observed for M1-corticostriatal afferents. Critically, in vivo optogenetic stimulation of S1-corticostriatal afferents produced task-specific behavioral inhibition, which was bidirectionally modulated by striatal PV interneurons. Optogenetic stimulation of M1 afferents produced the opposite behavioral effect. Thus, our results suggest opposing roles for sensory and motor cortex in behavioral choice via distinct influences on striatal circuitry.


Subject(s)
Choice Behavior/physiology , Corpus Striatum/physiology , Motor Cortex/physiology , Somatosensory Cortex/physiology , Animals , Female , Interneurons/physiology , Male , Mice , Neural Pathways/physiology
SELECTION OF CITATIONS
SEARCH DETAIL