Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(7): 1549-1560.e3, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38458192

ABSTRACT

The successful pursuit of goals requires the coordinated execution and termination of actions that lead to positive outcomes. This process relies on motivational states that are guided by internal drivers, such as hunger or fear. However, the mechanisms by which the brain tracks motivational states to shape instrumental actions are not fully understood. The paraventricular nucleus of the thalamus (PVT) is a midline thalamic nucleus that shapes motivated behaviors via its projections to the nucleus accumbens (NAc)1,2,3,4,5,6,7,8 and monitors internal state via interoceptive inputs from the hypothalamus and brainstem.3,9,10,11,12,13,14 Recent studies indicate that the PVT can be subdivided into two major neuronal subpopulations, namely PVTD2(+) and PVTD2(-), which differ in genetic identity, functionality, and anatomical connectivity to other brain regions, including the NAc.4,15,16 In this study, we used fiber photometry to investigate the in vivo dynamics of these two distinct PVT neuronal types in mice performing a foraging-like behavioral task. We discovered that PVTD2(+) and PVTD2(-) neurons encode the execution and termination of goal-oriented actions, respectively. Furthermore, activity in the PVTD2(+) neuronal population mirrored motivation parameters such as vigor and satiety. Similarly, PVTD2(-) neurons also mirrored some of these parameters, but to a much lesser extent. Importantly, these features were largely preserved when activity in PVT projections to the NAc was selectively assessed. Collectively, our results highlight the existence of two parallel thalamo-striatal projections that participate in the dynamic regulation of goal pursuits and provide insight into the mechanisms by which the brain tracks motivational states to shape instrumental actions.


Subject(s)
Motivation , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/physiology , Thalamus , Midline Thalamic Nuclei/physiology , Hypothalamus
2.
Nature ; 626(8001): 1066-1072, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326610

ABSTRACT

Animals can learn about sources of danger while minimizing their own risk by observing how others respond to threats. However, the distinct neural mechanisms by which threats are learned through social observation (known as observational fear learning1-4 (OFL)) to generate behavioural responses specific to such threats remain poorly understood. The dorsomedial prefrontal cortex (dmPFC) performs several key functions that may underlie OFL, including processing of social information and disambiguation of threat cues5-11. Here we show that dmPFC is recruited and required for OFL in mice. Using cellular-resolution microendoscopic calcium imaging, we demonstrate that dmPFC neurons code for observational fear and do so in a manner that is distinct from direct experience. We find that dmPFC neuronal activity predicts upcoming switches between freezing and moving state elicited by threat. By combining neuronal circuit mapping, calcium imaging, electrophysiological recordings and optogenetics, we show that dmPFC projections to the midbrain periaqueductal grey (PAG) constrain observer freezing, and that amygdalar and hippocampal inputs to dmPFC opposingly modulate observer freezing. Together our findings reveal that dmPFC neurons compute a distinct code for observational fear and coordinate long-range neural circuits to select behavioural responses.


Subject(s)
Cues , Fear , Neural Pathways , Prefrontal Cortex , Social Learning , Animals , Mice , Amygdala/physiology , Calcium/metabolism , Electrophysiology , Fear/physiology , Hippocampus/physiology , Neural Pathways/physiology , Neurons/physiology , Optogenetics , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Photic Stimulation , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Social Learning/physiology , Freezing Reaction, Cataleptic/physiology
3.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-37781624

ABSTRACT

The successful pursuit of goals requires the coordinated execution and termination of actions that lead to positive outcomes. This process is thought to rely on motivational states that are guided by internal drivers, such as hunger or fear. However, the mechanisms by which the brain tracks motivational states to shape instrumental actions are not fully understood. The paraventricular nucleus of the thalamus (PVT) is a midline thalamic nucleus that shapes motivated behaviors via its projections to the nucleus accumbens (NAc)1-8 and monitors internal state via interoceptive inputs from the hypothalamus and brainstem3,9-14. Recent studies indicate that the PVT can be subdivided into two major neuronal subpopulations, namely PVTD2(+) and PVTD2(-), which differ in genetic identity, functionality, and anatomical connectivity to other brain regions, including the NAc4,15,16. In this study, we used fiber photometry to investigate the in vivo dynamics of these two distinct PVT neuronal types in mice performing a reward foraging-like behavioral task. We discovered that PVTD2(+) and PVTD2(-) neurons encode the execution and termination of goal-oriented actions, respectively. Furthermore, activity in the PVTD2(+) neuronal population mirrored motivation parameters such as vigor and satiety. Similarly, PVTD2(-) neurons, also mirrored some of these parameters but to a much lesser extent. Importantly, these features were largely preserved when activity in PVT projections to the NAc was selectively assessed. Collectively, our results highlight the existence of two parallel thalamo-striatal projections that participate in the dynamic regulation of goal pursuits and provide insight into the mechanisms by which the brain tracks motivational states to shape instrumental actions.

4.
Trends Neurosci ; 46(9): 701-711, 2023 09.
Article in English | MEDLINE | ID: mdl-37495461

ABSTRACT

Plasticity elicited by fear conditioning (FC) is thought to support the storage of aversive associative memories. Although work over the past decade has revealed FC-induced plasticity beyond canonical sites in the basolateral complex of the amygdala (BLA), it is not known whether modifications across distributed circuits make equivalent or distinct contributions to aversive memory. Here, we review evidence demonstrating that experience-dependent synaptic plasticity in the central nucleus of the amygdala (CeA) has a circumscribed role in memory expression per se, guiding the selection of defensive programs in response to acquired threats. We argue that the CeA may be a key example of a broader phenomenon by which synaptic plasticity at specific nodes of a distributed network makes a complementary contribution to distinct memory processes.


Subject(s)
Central Amygdaloid Nucleus , Humans , Fear/physiology , Neuronal Plasticity/physiology , Memory/physiology , Affect
5.
Elife ; 122023 03 03.
Article in English | MEDLINE | ID: mdl-36867023

ABSTRACT

The paraventricular nucleus of the thalamus (PVT) is known to regulate various cognitive and behavioral processes. However, while functional diversity among PVT circuits has often been linked to cellular differences, the molecular identity and spatial distribution of PVT cell types remain unclear. To address this gap, here we used single nucleus RNA sequencing (snRNA-seq) and identified five molecularly distinct PVT neuronal subtypes in the mouse brain. Additionally, multiplex fluorescent in situ hybridization of top marker genes revealed that PVT subtypes are organized by a combination of previously unidentified molecular gradients. Lastly, comparing our dataset with a recently published single-cell sequencing atlas of the thalamus yielded novel insight into the PVT's connectivity with the cortex, including unexpected innervation of auditory and visual areas. This comparison also revealed that our data contains a largely non-overlapping transcriptomic map of multiple midline thalamic nuclei. Collectively, our findings uncover previously unknown features of the molecular diversity and anatomical organization of the PVT and provide a valuable resource for future investigations.


Subject(s)
Paraventricular Hypothalamic Nucleus , Thalamus , Rats , Mice , Animals , In Situ Hybridization, Fluorescence , Rats, Sprague-Dawley , Neural Pathways/physiology , Midline Thalamic Nuclei/metabolism
6.
Nat Neurosci ; 26(4): 594-605, 2023 04.
Article in English | MEDLINE | ID: mdl-36894654

ABSTRACT

Supraspinal brain regions modify nociceptive signals in response to various stressors including stimuli that elevate pain thresholds. The medulla oblongata has previously been implicated in this type of pain control, but the neurons and molecular circuits involved have remained elusive. Here we identify catecholaminergic neurons in the caudal ventrolateral medulla that are activated by noxious stimuli in mice. Upon activation, these neurons produce bilateral feed-forward inhibition that attenuates nociceptive responses through a pathway involving the locus coeruleus and norepinephrine in the spinal cord. This pathway is sufficient to attenuate injury-induced heat allodynia and is required for counter-stimulus induced analgesia to noxious heat. Our findings define a component of the pain modulatory system that regulates nociceptive responses.


Subject(s)
Nociceptors , Pain , Mice , Animals , Nociceptors/physiology , Pain/metabolism , Medulla Oblongata/metabolism , Pain Management , Neurons/physiology , Spinal Cord/physiology
7.
Elife ; 112022 10 21.
Article in English | MEDLINE | ID: mdl-36269044

ABSTRACT

Central amygdala neurons expressing protein kinase C-delta (CeA-PKCδ) are sensitized following nerve injury and promote pain-related responses in mice. The neural circuits underlying modulation of pain-related behaviors by CeA-PKCδ neurons, however, remain unknown. In this study, we identified a neural circuit that originates in CeA-PKCδ neurons and terminates in the ventral region of the zona incerta (ZI), a subthalamic structure previously linked to pain processing. Behavioral experiments show that chemogenetic inhibition of GABAergic ZI neurons induced bilateral hypersensitivity in uninjured mice and contralateral hypersensitivity after nerve injury. In contrast, chemogenetic activation of GABAergic ZI neurons reversed nerve injury-induced hypersensitivity. Optogenetic manipulations of CeA-PKCδ axonal terminals in the ZI further showed that inhibition of this pathway reduces nerve injury-induced hypersensitivity whereas activation of the pathway produces hypersensitivity in the uninjured paws. Altogether, our results identify a novel nociceptive inhibitory efferent pathway from CeA-PKCδ neurons to the ZI that bidirectionally modulates pain-related behaviors in mice.


Subject(s)
Central Amygdaloid Nucleus , Zona Incerta , Animals , Mice , Zona Incerta/physiology , Pain , GABAergic Neurons/physiology , Optogenetics
8.
Nat Neurosci ; 25(8): 999-1008, 2022 08.
Article in English | MEDLINE | ID: mdl-35915178

ABSTRACT

In nature, animals display defensive behaviors that reflect the spatiotemporal distance of threats. Laboratory-based paradigms that elicit specific defensive responses in rodents have provided valuable insight into the brain mechanisms that mediate the construction of defensive modes with varying degrees of threat imminence. In this Review, we discuss accumulating evidence that the central nucleus of the amygdala (CeA) plays a key role in this process. Specifically, we propose that the mutually inhibitory circuits of the CeA use a winner-takes-all strategy that supports transitioning across defensive modes and the execution of specific defensive behaviors to previously formed threat associations. Our proposal provides a conceptual framework in which seemingly divergent observations regarding CeA function can be interpreted and identifies various areas of priority for future research.


Subject(s)
Central Amygdaloid Nucleus , Animals , Central Amygdaloid Nucleus/physiology
9.
Nat Neurosci ; 24(10): 1429-1440, 2021 10.
Article in English | MEDLINE | ID: mdl-34413514

ABSTRACT

The appropriate selection of passive and active defensive behaviors in threatening situations is essential for survival. Previous studies have shown that passive defensive responses depend on activity of the central nucleus of the amygdala (CeA), whereas active ones primarily rely on the nucleus accumbens (NAc). However, the mechanisms underlying flexible switching between these two types of responses remain unknown. Here we show in mice that the paraventricular thalamus (PVT) mediates the selection of defensive behaviors through its interaction with the CeA and the NAc. We show that the PVT-CeA pathway drives conditioned freezing responses, whereas the PVT-NAc pathway is inhibited during freezing and, instead, signals active avoidance events. Optogenetic manipulations revealed that activity in the PVT-CeA or PVT-NAc pathway biases behavior toward the selection of passive or active defensive responses, respectively. These findings provide evidence that the PVT mediates flexible switching between opposing defensive behaviors.


Subject(s)
Behavior, Animal/physiology , Neural Pathways/physiology , Paraventricular Hypothalamic Nucleus/physiology , Animals , Avoidance Learning , Mice , Mice, Inbred C57BL , Optogenetics
10.
Trends Neurosci ; 44(7): 538-549, 2021 07.
Article in English | MEDLINE | ID: mdl-33775435

ABSTRACT

Early anatomical evidence suggested that the paraventricular nucleus of the thalamus (PVT) regulates arousal, as well as emotional and motivated behaviors. We discuss recent studies using modern techniques which now confirm and expand the involvement of the rodent PVT in these functions. Despite the emerging notion that the PVT is implicated in various behavioral processes, a recurrent theme is that activity in this brain region depends on internal state information arriving from the hypothalamus and brainstem, and is influenced by prior experience. We propose that the primary function of the PVT is to detect homeostatic challenges by integrating information about prior experiences, competing needs, and internal state to guide adaptive behavioral responses aimed at restoring homeostasis.


Subject(s)
Midline Thalamic Nuclei , Paraventricular Hypothalamic Nucleus , Homeostasis , Humans , Neurons , Thalamus
11.
Nat Commun ; 11(1): 6218, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277492

ABSTRACT

Marked deficits in glucose availability, or glucoprivation, elicit organism-wide counter-regulatory responses whose purpose is to restore glucose homeostasis. However, while catecholamine neurons of the ventrolateral medulla (VLMCA) are thought to orchestrate these responses, the circuit and cellular mechanisms underlying specific counter-regulatory responses are largely unknown. Here, we combined anatomical, imaging, optogenetic and behavioral approaches to interrogate the circuit mechanisms by which VLMCA neurons orchestrate glucoprivation-induced food seeking behavior. Using these approaches, we found that VLMCA neurons form functional connections with nucleus accumbens (NAc)-projecting neurons of the posterior portion of the paraventricular nucleus of the thalamus (pPVT). Importantly, optogenetic manipulations revealed that while activation of VLMCA projections to the pPVT was sufficient to elicit robust feeding behavior in well fed mice, inhibition of VLMCA-pPVT communication significantly impaired glucoprivation-induced feeding while leaving other major counterregulatory responses intact. Collectively our findings identify the VLMCA-pPVT-NAc pathway as a previously-neglected node selectively controlling glucoprivation-induced food seeking. Moreover, by identifying the ventrolateral medulla as a direct source of metabolic information to the midline thalamus, our results support a growing body of literature on the role of the PVT in homeostatic regulation.


Subject(s)
Catecholamines/metabolism , Feeding Behavior/physiology , Glucose/metabolism , Medulla Oblongata/physiology , Neurons/physiology , Ventral Thalamic Nuclei/physiology , Animals , Female , Homeostasis/physiology , Male , Medulla Oblongata/cytology , Mice, Inbred C57BL , Mice, Transgenic , Midline Thalamic Nuclei/cytology , Midline Thalamic Nuclei/physiology , Neurons/metabolism , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Ventral Thalamic Nuclei/cytology
12.
Nature ; 581(7807): 194-198, 2020 05.
Article in English | MEDLINE | ID: mdl-32404998

ABSTRACT

Daily changes in light and food availability are major time cues that influence circadian timing1. However, little is known about the circuits that integrate these time cues to drive a coherent circadian output1-3. Here we investigate whether retinal inputs modulate entrainment to nonphotic cues such as time-restricted feeding. Photic information is relayed to the suprachiasmatic nucleus (SCN)-the central circadian pacemaker-and the intergeniculate leaflet (IGL) through intrinsically photosensitive retinal ganglion cells (ipRGCs)4. We show that adult mice that lack ipRGCs from the early postnatal stages have impaired entrainment to time-restricted feeding, whereas ablation of ipRGCs at later stages had no effect. Innervation of ipRGCs at early postnatal stages influences IGL neurons that express neuropeptide Y (NPY) (hereafter, IGLNPY neurons), guiding the assembly of a functional IGLNPY-SCN circuit. Moreover, silencing IGLNPY neurons in adult mice mimicked the deficits that were induced by ablation of ipRGCs in the early postnatal stages, and acute inhibition of IGLNPY terminals in the SCN decreased food-anticipatory activity. Thus, innervation of ipRGCs in the early postnatal period tunes the IGLNPY-SCN circuit to allow entrainment to time-restricted feeding.


Subject(s)
Circadian Rhythm/physiology , Feeding Behavior/physiology , Light , Neural Pathways , Retina/physiology , Animals , Axons/physiology , Axons/radiation effects , Circadian Rhythm/radiation effects , Cues , Eating/physiology , Eating/radiation effects , Feeding Behavior/radiation effects , Female , Geniculate Bodies/cytology , Geniculate Bodies/physiology , Geniculate Bodies/radiation effects , Male , Mice , Neural Pathways/radiation effects , Neuropeptide Y/metabolism , Retina/cytology , Retina/radiation effects , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Signal Transduction/radiation effects , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology , Suprachiasmatic Nucleus/radiation effects , Time Factors
13.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32029441

ABSTRACT

Altered cortical excitation-inhibition (E-I) balance resulting from abnormal parvalbumin interneuron (PV IN) function is a proposed pathophysiological mechanism of schizophrenia and other major psychiatric disorders. Preclinical studies have indicated that disrupted-in-schizophrenia-1 (Disc1) is a useful molecular lead to address the biology of prefrontal cortex (PFC)-dependent cognition and PV IN function. To date, PFC inhibitory circuit function has not been investigated in depth in Disc1 locus impairment (LI) mouse models. Therefore, we used a Disc1 LI mouse model to investigate E-I balance in medial PFC (mPFC) circuits. We found that inhibition onto layer 2/3 excitatory pyramidal neurons in the mPFC was significantly reduced in Disc1 LI mice. This reduced inhibition was accompanied by decreased GABA release from local PV, but not somatostatin (SOM) INs, and by impaired feedforward inhibition (FFI) in the mediodorsal thalamus (MD) to mPFC circuit. Our mechanistic findings of abnormal PV IN function in a Disc1 LI model provide insight into biology that may be relevant to neuropsychiatric disorders including schizophrenia.


Subject(s)
Interneurons , Parvalbumins , Animals , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Somatostatin
14.
Nat Neurosci ; 23(2): 217-228, 2020 02.
Article in English | MEDLINE | ID: mdl-31932767

ABSTRACT

Unlike the sensory thalamus, studies on the functional organization of the midline and intralaminar nuclei are scarce, and this has hindered the establishment of conceptual models of the function of this brain region. We investigated the functional organization of the paraventricular nucleus of the thalamus (PVT), a midline thalamic structure that is increasingly being recognized as a critical node in the control of diverse processes such as arousal, stress, emotional memory and motivation, in mice. We identify two major classes of PVT neurons-termed type I and type II-that differ in terms of gene expression, anatomy and function. In addition, we demonstrate that type II neurons belong to a previously neglected class of PVT neurons that convey arousal-related information to corticothalamic neurons of the infralimbic cortex. Our results uncover the existence of an arousal-modulated thalamo-corticothalamic loop that links the PVT and the ventromedial prefrontal cortex.


Subject(s)
Neurons/cytology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/physiology
15.
Nat Neurosci ; 21(7): 963-973, 2018 07.
Article in English | MEDLINE | ID: mdl-29915192

ABSTRACT

The paraventricular nucleus of the thalamus (PVT) is increasingly being recognized as a critical node linking stress detection to the emergence of adaptive behavioral responses to stress. However, despite growing evidence implicating the PVT in stress processing, the neural mechanisms by which stress impacts PVT neurocircuitry and promotes stressed states remain unknown. Here we show that stress exposure drives a rapid and persistent reduction of inhibitory transmission onto projection neurons of the posterior PVT (pPVT). This stress-induced disinhibition of the pPVT was associated with a locus coeruleus-mediated rise in the extracellular concentration of dopamine in the midline thalamus, required the function of dopamine D2 receptors on PVT neurons, and increased sensitivity to stress. Our findings define the locus coeruleus as an important modulator of PVT function: by controlling the inhibitory tone of the pPVT, it modulates the excitability of pPVT projection neurons and controls stress responsivity.


Subject(s)
Dopamine/metabolism , Locus Coeruleus/metabolism , Neural Inhibition/physiology , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Stress, Physiological/physiology , Thalamus/metabolism , Animals , Electroshock , Fear/physiology , Female , Membrane Glycoproteins , Mice , Neural Pathways , Receptors, Interleukin-1
16.
J Neurosci ; 38(24): 5567-5583, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29844022

ABSTRACT

Both the amygdala and the bed nucleus of the stria terminalis (BNST) have been implicated in maladaptive anxiety characteristics of anxiety disorders. However, the underlying circuit and cellular mechanisms have remained elusive. Here we show that mice with Erbb4 gene deficiency in somatostatin-expressing (SOM+) neurons exhibit heightened anxiety as measured in the elevated plus maze test and the open field test, two assays commonly used to assess anxiety-related behaviors in rodents. Using a combination of electrophysiological, molecular, genetic, and pharmacological techniques, we demonstrate that the abnormal anxiety in the mutant mice is caused by enhanced excitatory synaptic inputs onto SOM+ neurons in the central amygdala (CeA), and the resulting reduction in inhibition onto downstream SOM+ neurons in the BNST. Notably, our results indicate that an increase in dynorphin signaling in SOM+ CeA neurons mediates the paradoxical reduction in inhibition onto SOM+ BNST neurons, and that the consequent enhanced activity of SOM+ BNST neurons is both necessary for and sufficient to drive the elevated anxiety. Finally, we show that the elevated anxiety and the associated synaptic dysfunctions and increased dynorphin signaling in the CeA-BNST circuit of the Erbb4 mutant mice can be recapitulated by stress in wild-type mice. Together, our results unravel previously unknown circuit and cellular processes in the central extended amygdala that can cause maladaptive anxiety.SIGNIFICANCE STATEMENT The central extended amygdala has been implicated in anxiety-related behaviors, but the underlying mechanisms are unclear. Here we found that somatostatin-expressing neurons in the central amygdala (CeA) controls anxiety through modulation of the stria terminalis, a process that is mediated by an increase in dynorphin signaling in the CeA. Our results reveal circuit and cellular dysfunctions that may account for maladaptive anxiety.


Subject(s)
Anxiety/physiopathology , Central Amygdaloid Nucleus/physiopathology , Neural Pathways/physiology , Septal Nuclei/physiopathology , Animals , Central Amygdaloid Nucleus/metabolism , Dynorphins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Receptor, ErbB-4/deficiency , Septal Nuclei/metabolism , Somatostatin/metabolism
17.
J Neurosci ; 38(6): 1418-1429, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29305535

ABSTRACT

For animals to survive, they must reliably predict during foraging which substances are suitable for consumption. Despite extensive study, the neural circuit mechanisms underlying such adaptive behavior remain poorly understood. Here, using a tastant (sucrose/quinine)-reinforced "go/no-go" task in male and female mice, we examined the anatomical and functional connectivity of the circuit linking the insular cortex (IC) to the central amygdala (CeA) and the role of this circuit in the establishment of appropriate behavioral responses. Using anatomic tracing approaches combined with optogenetics-assisted circuit mapping, we found that the gustatory region of the IC sends direct excitatory projections to the lateral division of the CeA (CeL), making monosynaptic excitatory connections with distinct populations of CeL neurons. Specific inhibition of neurotransmitter release from the CeL-projecting IC neurons prevented mice from acquiring the "no-go" response, and impaired the "go" responses in the go/no-go task. Furthermore, selective activation of the IC-CeL pathway with optogenetics drove unconditioned lick suppression in thirsty animals, induced aversive responses, and was sufficient to instruct conditioned action suppression in response to a cue predicting the optogenetic activation. These results indicate that activities in the IC-CeL circuit are critical for establishing taste-reinforced behavioral responses, including avoidance responses to an aversive tastant, and are sufficient to drive learning of anticipatory avoidance. Our findings suggest that the IC-CeL circuit plays an important role in guiding appropriate choices during foraging.SIGNIFICANCE STATEMENT An animal's ability to predict which substances are suitable for consumption and then produce an appropriate action to those substances is critical for survival. Here we found that activity in the circuit that links the insular cortex (IC) to the central amygdala (CeA) is necessary for establishing appropriate behavioral responses to taste-predicting cues. This neural circuit seems to be particularly tuned to avoid an unpleasant tastant, and is also sufficient to drive learning of such avoidance responses. These results suggest that the IC-CeA circuit is critical for generating appropriate behavioral responses during foraging when facing different choices.


Subject(s)
Central Amygdaloid Nucleus/drug effects , Cerebral Cortex/drug effects , Choice Behavior/drug effects , Flavoring Agents/pharmacology , Neural Pathways/drug effects , Animals , Avoidance Learning/drug effects , Brain Mapping , Conditioning, Operant/drug effects , Cues , Female , Male , Mice , Mice, Inbred C57BL , Optogenetics , Quinine/pharmacology , Sucrose/pharmacology , Sweetening Agents/pharmacology , Synaptic Transmission/drug effects , Taste/drug effects , Thirst/physiology
18.
Nature ; 539(7628): 289-293, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27652894

ABSTRACT

The basal ganglia, a group of subcortical nuclei, play a crucial role in decision-making by selecting actions and evaluating their outcomes. While much is known about the function of the basal ganglia circuitry in selection, how these nuclei contribute to outcome evaluation is less clear. Here we show that neurons in the habenula-projecting globus pallidus (GPh) in mice are essential for evaluating action outcomes and are regulated by a specific set of inputs from the basal ganglia. We find in a classical conditioning task that individual mouse GPh neurons bidirectionally encode whether an outcome is better or worse than expected. Mimicking these evaluation signals with optogenetic inhibition or excitation is sufficient to reinforce or discourage actions in a decision-making task. Moreover, cell-type-specific synaptic manipulations reveal that the inhibitory and excitatory inputs to the GPh are necessary for mice to appropriately evaluate positive and negative feedback, respectively. Finally, using rabies-virus-assisted monosynaptic tracing, we show that the GPh is embedded in a basal ganglia circuit wherein it receives inhibitory input from both striosomal and matrix compartments of the striatum, and excitatory input from the 'limbic' regions of the subthalamic nucleus. Our results provide evidence that information about the selection and evaluation of actions is channelled through distinct sets of basal ganglia circuits, with the GPh representing a key locus in which information of opposing valence is integrated to determine whether action outcomes are better or worse than expected.


Subject(s)
Basal Ganglia/cytology , Basal Ganglia/physiology , Decision Making , Neural Pathways/physiology , Punishment , Reward , Animals , Conditioning, Classical , Feedback, Physiological , Female , Globus Pallidus/cytology , Globus Pallidus/physiology , Glutamic Acid/metabolism , Habenula/cytology , Habenula/physiology , Male , Mice , Neurons/metabolism , Optogenetics , Rabies virus/physiology , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
19.
Nature ; 519(7544): 455-9, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25600269

ABSTRACT

Appropriate responses to an imminent threat brace us for adversities. The ability to sense and predict threatening or stressful events is essential for such adaptive behaviour. In the mammalian brain, one putative stress sensor is the paraventricular nucleus of the thalamus (PVT), an area that is readily activated by both physical and psychological stressors. However, the role of the PVT in the establishment of adaptive behavioural responses remains unclear. Here we show in mice that the PVT regulates fear processing in the lateral division of the central amygdala (CeL), a structure that orchestrates fear learning and expression. Selective inactivation of CeL-projecting PVT neurons prevented fear conditioning, an effect that can be accounted for by an impairment in fear-conditioning-induced synaptic potentiation onto somatostatin-expressing (SOM(+)) CeL neurons, which has previously been shown to store fear memory. Consistently, we found that PVT neurons preferentially innervate SOM(+) neurons in the CeL, and stimulation of PVT afferents facilitated SOM(+) neuron activity and promoted intra-CeL inhibition, two processes that are critical for fear learning and expression. Notably, PVT modulation of SOM(+) CeL neurons was mediated by activation of the brain-derived neurotrophic factor (BDNF) receptor tropomysin-related kinase B (TrkB). As a result, selective deletion of either Bdnf in the PVT or Trkb in SOM(+) CeL neurons impaired fear conditioning, while infusion of BDNF into the CeL enhanced fear learning and elicited unconditioned fear responses. Our results demonstrate that the PVT-CeL pathway constitutes a novel circuit essential for both the establishment of fear memory and the expression of fear responses, and uncover mechanisms linking stress detection in PVT with the emergence of adaptive behaviour.


Subject(s)
Central Amygdaloid Nucleus/physiology , Fear/physiology , Neural Pathways/physiology , Thalamus/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Central Amygdaloid Nucleus/cytology , Conditioning, Psychological/physiology , Fear/psychology , Female , Male , Memory/physiology , Mice , Neural Pathways/cytology , Neuronal Plasticity , Neurons/metabolism , Receptor, trkB/metabolism , Somatostatin/metabolism , Thalamus/cytology , Time Factors
20.
J Neurosci ; 34(7): 2432-7, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24523533

ABSTRACT

Recent studies indicate that the lateral subdivision of the central amygdala (CeL) is essential for fear learning. Specifically, fear conditioning induces cell-type-specific synaptic plasticity in CeL neurons that is required for the storage of fear memories. The CeL also controls fear expression by gating the activity of the medial subdivision of the central amygdala (CeM), the canonical amygdala output to areas that mediate defensive responses. In addition to the connection with CeM, the CeL sends long-range projections to innervate extra-amygdala areas. However, the long-range projection CeL neurons have not been well characterized, and their role in fear regulation is unknown. Here we show in mice that a subset of CeL neurons directly project to the midbrain periaqueductal gray (PAG) and the paraventricular nucleus of the thalamus, two brain areas implicated in defensive behavior. These long-range projection CeL neurons are predominantly somatostatin-positive (SOM(+)) neurons, which can directly inhibit PAG neurons, and some of which innervate both the PAG and paraventricular nucleus of the thalamus. Notably, fear conditioning potentiates excitatory synaptic transmission onto these long-range projection CeL neurons. Thus, our study identifies a subpopulation of SOM(+) CeL neurons that may contribute to fear learning and regulate fear expression independent of CeM.


Subject(s)
Amygdala/cytology , Fear/physiology , Neurons/cytology , Synaptic Transmission/physiology , Amygdala/metabolism , Animals , Conditioning, Classical/physiology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...