Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Front Neuroanat ; 12: 91, 2018.
Article in English | MEDLINE | ID: mdl-30467465

ABSTRACT

Our original review, "Heterogeneity and Diversity of Striatal GABAergic Interneurons," to which this is an invited update, was published in December, 2010 in Frontiers is Neuroanatomy. In that article, we reviewed several decades' worth of anatomical and electrophysiological data on striatal parvalbumin (PV)-, neuropeptide Y (NPY)- and calretinin(CR)-expressing GABAergic interneurons from many laboratories including our own. In addition, we reported on a recently discovered novel tyrosine hydroxylase (TH) expressing GABAergic interneuron class first revealed in transgenic TH EGFP reporter mouse line. In this review, we report on further advances in the understanding of the functional properties of previously reported striatal GABAergic interneurons and their synaptic connections. With the application of new transgenic fluorescent reporter and Cre-driver/reporter lines, plus optogenetic, chemogenetic and viral transduction methods, several additional subtypes of novel striatal GABAergic interneurons have been discovered, as well as the synaptic networks in which they are embedded. These findings make it clear that previous hypotheses in which striatal GABAergic interneurons modulate and/or control the firing of spiny neurons principally by simple feedforward and/or feedback inhibition are at best incomplete. A more accurate picture is one in which there are highly selective and specific afferent inputs, synaptic connections between different interneuron subtypes and spiny neurons and among different GABAergic interneurons that result in the formation of functional networks and ensembles of spiny neurons.

10.
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
11.
Nat Commun ; 8: 15860, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28604688

ABSTRACT

Recent discoveries of striatal GABAergic interneurons require a new conceptualization of the organization of intrastriatal circuitry and their cortical and thalamic inputs. We investigated thalamic inputs to the two populations of striatal neuropeptide Y (NPY) interneurons, plateau low threshold spike (PLTS) and NPY-neurogliaform (NGF) cells. Optogenetic activation of parafascicular inputs evokes suprathreshold monosynaptic glutamatergic excitation in NGF interneurons and a disynaptic, nicotinic excitation through cholinergic interneurons. In contrast, the predominant response of PLTS interneurons is a disynaptic inhibition dependent on thalamic activation of striatal tyrosine hydroxylase interneurons (THINs). In contrast, THINs do not innervate NGF or fast spiking interneurons, showing significant specificity in THINs outputs. Chemospecific ablation of THINs impairs prepulse inhibition of the acoustic startle response suggesting an important behavioural role of this disynaptic pathway. Our findings demonstrate that the impact of the parafascicular nucleus on striatal activity and some related behaviour critically depend on synaptic interactions within interneuronal circuits.


Subject(s)
GABAergic Neurons/physiology , Interneurons/physiology , Neuropeptide Y/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiology , Interneurons/metabolism , Mice , Mice, Transgenic , Optogenetics , Synaptic Transmission , Thalamus , Tyrosine 3-Monooxygenase/metabolism
12.
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
13.
Nat Neurosci ; 19(8): 1025-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27348215

ABSTRACT

Dopamine neurons in the ventral tegmental area (VTA) receive cholinergic innervation from brainstem structures that are associated with either movement or reward. Whereas cholinergic neurons of the pedunculopontine nucleus (PPN) carry an associative/motor signal, those of the laterodorsal tegmental nucleus (LDT) convey limbic information. We used optogenetics and in vivo juxtacellular recording and labeling to examine the influence of brainstem cholinergic innervation of distinct neuronal subpopulations in the VTA. We found that LDT cholinergic axons selectively enhanced the bursting activity of mesolimbic dopamine neurons that were excited by aversive stimulation. In contrast, PPN cholinergic axons activated and changed the discharge properties of VTA neurons that were integrated in distinct functional circuits and were inhibited by aversive stimulation. Although both structures conveyed a reinforcing signal, they had opposite roles in locomotion. Our results demonstrate that two modes of cholinergic transmission operate in the VTA and segregate the neurons involved in different reward circuits.


Subject(s)
Acetylcholine/pharmacology , Neural Pathways/drug effects , Neurons/drug effects , Nucleus Accumbens/drug effects , Ventral Tegmental Area/drug effects , Animals , Cholinergic Agents/pharmacology , Dopamine/pharmacology , Dopaminergic Neurons/drug effects , Male , Neural Pathways/physiology , Nucleus Accumbens/metabolism , Rats, Long-Evans , Tegmentum Mesencephali/drug effects , Ventral Tegmental Area/physiology
14.
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
15.
Neuropharmacology ; 95: 468-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25908399

ABSTRACT

The recent electrophysiological characterization of TH-expressing GABAergic interneurons (THINs) in the neostriatum revealed an unexpected degree of diversity of interneurons in this brain area (Ibáñez-Sandoval et al., 2010, Unal et al., 2011, 2015). Despite being relatively few in number, THINs may play a significant role in transmitting and distributing extra- and intrastriatal neuromodulatory signals in the striatal circuitry. Here we investigated the dopaminergic and cholinergic regulation of THINs in vitro. We found that the dominant effect of dopamine was a dramatic enhancement of the ability of THINs to generate long-lasting depolarizing plateau potentials (PPs). Interestingly, the same effect could also be elicited by amphetamine-induced release of endogenous dopamine suggesting that THINs may exhibit similar responses to changes in extracellular dopamine concentration in vivo. The enhancement of PPs in THINs is perhaps the most pronounced effect of dopamine on the intrinsic excitability of neostriatal neurons described to date. Further, we demonstrate that all subtypes of THINSs tested also express nicotinic cholinergic receptors. All THIS responded, albeit differentially, with depolarization, PPs and spiking to brief application of nicotinic agonists. Powerful modulation of the nonlinear integrative properties of THINs by dopamine and the direct depolarization of these neurons by acetylcholine may play important roles in mediating the effects of these neuromodulators in the neostriatum with potentially important implications for understanding the mechanisms of neuropsychiatric disorders affecting the basal ganglia.


Subject(s)
Acetylcholine/metabolism , Dopamine/metabolism , Interneurons/physiology , Neostriatum/physiology , Tyrosine 3-Monooxygenase/metabolism , Animals , Electric Stimulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interneurons/cytology , Interneurons/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Transgenic , Neostriatum/cytology , Neostriatum/drug effects , Nicotinic Agonists/pharmacology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D5/agonists , Receptors, Dopamine D5/metabolism , Receptors, Nicotinic/metabolism , Tissue Culture Techniques , Tyrosine 3-Monooxygenase/genetics
16.
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
17.
Brain Struct Funct ; 220(1): 331-49, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24173616

ABSTRACT

Using transgenic mice that express enhanced green fluorescent protein (EGFP) under the control of the tyrosine hydroxylase (TH) promoter, we have previously shown that there are approximately 3,000 striatal EGFP-TH interneurons per hemisphere in mice. Here, we report that striatal TH-EGFP interneurons exhibit a small, transient but significant increase in number after unilateral destruction of the nigrostriatal dopaminergic pathway. The increase in cell number is accompanied by electrophysiological and morphological changes. The intrinsic electrophysiological properties of EGFP-TH interneurons ipsilateral to 6-OHDA lesion were similar to those originally reported in intact mice except for a significant reduction in the duration of a characteristic depolarization induced plateau potential. There was a significant change in the distribution of the four previously described electrophysiologically distinct subtypes of striatal TH interneurons. There was a concomitant increase in the frequency of both spontaneous excitatory and inhibitory post-synaptic currents, while their amplitudes did not change. Nigrostriatal lesions did not affect somatic size or dendritic length or branching, but resulted in an increase in the density of proximal dendritic spines and spine-like appendages in EGFP-TH interneurons. The changes indicate that electrophysiology properties and morphology of striatal EGFP-TH interneurons depend on endogenous levels of dopamine arising from the nigrostriatal pathway. Furthermore, these changes may serve to help compensate for the changes in activity of spiny projection neurons that occur following loss of the nigrostriatal innervation in experimental or in early idiopathic Parkinson's disease by increasing feedforward GABAergic inhibition exerted by these interneurons.


Subject(s)
Action Potentials/physiology , Corpus Striatum/cytology , Dopamine/metabolism , Neural Pathways/physiology , Neurons/physiology , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase/metabolism , Action Potentials/drug effects , Animals , Cell Count , Excitatory Amino Acid Antagonists/pharmacology , Female , Functional Laterality/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neuropeptide Y/metabolism , Oxidopamine/toxicity , Sympatholytics/toxicity , Time Factors , Tyrosine 3-Monooxygenase/genetics
18.
Cancer Res ; 74(17): 4845-4852, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25056119

ABSTRACT

Following mutations in BRAF and NRAS, the RAC1 c.85C>T single-nucleotide variant (SNV) encoding P29S amino acid change represents the next most frequently observed protein-coding hotspot mutation in melanoma. However, the biologic and clinical significance of the RAC1 P29S somatic mutation in approximately 4% to 9% of patients remains unclear. Here, we demonstrate that melanoma cell lines possessing the RAC1 hotspot variant are resistant to RAF inhibitors (vemurafenib and dabrafenib). Enforced expression of RAC1 P29S in sensitive BRAF-mutant melanoma cell lines confers resistance manifested by increased viability, decreased apoptosis, and enhanced tumor growth in vivo upon treatment with RAF inhibitors. Conversely, RNAi-mediated silencing of endogenous RAC1 P29S in a melanoma cell line with a co-occurring BRAF V600 mutation increased sensitivity to vemurafenib and dabrafenib. Our results suggest RAC1 P29S status may offer a predictive biomarker for RAF inhibitor resistance in melanoma patients, where it should be evaluated clinically.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , rac1 GTP-Binding Protein/genetics , raf Kinases/genetics , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Humans , Melanoma/drug therapy , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology
20.
Nat Neurosci ; 15(1): 123-30, 2011 Dec 11.
Article in English | MEDLINE | ID: mdl-22158514

ABSTRACT

Neostriatal cholinergic interneurons are believed to be important for reinforcement-mediated learning and response selection by signaling the occurrence and motivational value of behaviorally relevant stimuli through precisely timed multiphasic population responses. An important problem is to understand how these signals regulate the functioning of the neostriatum. Here we describe the synaptic organization of a previously unknown circuit that involves direct nicotinic excitation of several classes of GABAergic interneurons, including neuroptide Y-expressing neurogilaform neurons, and enables cholinergic interneurons to exert rapid inhibitory control of the activity of projection neurons. We also found that, in vivo, the dominant effect of an optogenetically reproduced pause-excitation population response of cholinergic interneurons was powerful and rapid inhibition of the firing of projection neurons that is coincident with synchronous cholinergic activation. These results reveal a previously unknown circuit mechanism that transmits reinforcement-related information of ChAT interneurons in the mouse neostriatal network.


Subject(s)
Acetylcholine/metabolism , Corpus Striatum/metabolism , Interneurons/metabolism , Nerve Net/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/physiology , Animals , Choline O-Acetyltransferase/metabolism , Inhibitory Postsynaptic Potentials/physiology , Mice , Mice, Transgenic , Neural Inhibition/physiology , Reinforcement, Psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...