Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Nutr Biochem ; 114: 109220, 2023 04.
Article in English | MEDLINE | ID: mdl-36435289

ABSTRACT

The incidence of adolescent mental health disorders is on the rise. Epidemiological studies suggest that poor nutrition is a significant contributor to this public health crisis, specifically through exposure to high level of dietary sugar, including fructose, during critical periods of development. Previous studies have shown that elevated fructose exposure during adolescence disrupts mental health. Despite these data, it is currently unknown how fructose exposure, specifically during infancy, may impact adolescent mental health. We developed a rat experimental protocol to investigate the effects of fructose exposure during infancy on behavioral, cognitive and metabolic endpoints in adolescence. We found that exposing rats to high fructose from birth to weaning resulted in higher circulating glucose, insulin and leptin levels in adolescence. High fructose during infancy also increased bodyweight, disrupted metabolic homeostasis in the basolateral amygdala (BLA) as indicated by decreased activity of the cellular energy sensor AMPK, and impaired attention and impulsivity in a male-specific manner. This impaired attention observed in adolescent male rats following neonatal fructose exposure was partially rescued by viral-mediated, in vivo expression of a constitutively active form of AMPK in principal neurons of the BLA. Our results suggest that exposure to high level of fructose during infancy may impact adolescent mental health in a male-specific manner and that manipulation of AMPK activity may mitigate this impact.


Subject(s)
Cognitive Dysfunction , Fructose , Female , Rats , Animals , Male , Fructose/adverse effects , AMP-Activated Protein Kinases/metabolism , Diet , Body Weight , Cognitive Dysfunction/etiology
2.
Brain Struct Funct ; 225(6): 1873-1888, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32556583

ABSTRACT

Recognizing reward-related stimuli is crucial for survival. Neuronal projections from the basolateral amygdala (BLA) to the nucleus accumbens (NAc) play an important role in processing reward-related cues. Previous studies revealed synchronization between distant brain regions in reward-sensitive neurocircuits; however, whether the NAc synchronizes with the BLA is unknown. Here, we recorded local field potentials simultaneously from the BLA and NAc of rats during social preference tests and an appetitive conditioning test in which explicit stimuli were associated with food. BLA-NAc coherence in the theta band (5-8 Hz) increased in response to food-associated cues. Meanwhile, the modulatory strength of theta-high gamma (50-110 Hz) phase-amplitude cross-frequency coupling (PAC) in the NAc decreased. Importantly, both of these neuromodulations disappeared upon extinction. In contrast, both theta and gamma power oscillations in each region increased in the presence of social conspecifics or contexts associated with conspecifics, but coherence did not change. To potentially disrupt behavior and associated neural activity, a subgroup of rats was exposed prenatally to valproic acid (VPA), which has been shown to disrupt transcriptome and excitatory/inhibitory balance in the amygdala. VPA-exposed rats demonstrated impulsive-like behavior, but VPA did not affect BLA-NAc coherence. These findings reveal changes in BLA-NAc coherence in response to select reward-related stimuli (i.e., food-predictive cues); the differences between the tasks used here could shed light onto the functional nature of BLA-NAc coherence and are discussed.


Subject(s)
Basolateral Nuclear Complex/physiology , Conditioning, Classical/physiology , Nucleus Accumbens/physiology , Reward , Animals , Appetitive Behavior/physiology , Brain Waves , Extinction, Psychological/physiology , Female , Neural Pathways/physiology , Rats, Sprague-Dawley , Social Behavior
3.
Neuropharmacology ; 150: 80-90, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30878403

ABSTRACT

Distinct regions and cell types in the anterolateral group of the bed nucleus of the stria terminalis (BNSTALG) act to modulate anxiety in opposing ways. A history of chronic stress increases anxiety-like behavior with lasting electrophysiological effects on the BNSTALG. However, the opposing circuits within the BNSTALG suggest that stress may have differential effects on the individual cell types that comprise these circuits to shift the balance to favor anxiogenesis. Yet, the effects of stress are generally examined by treating all neurons within a particular region of the BNST as a homogenoeus population. We used patch-clamp electrophysiology and single-cell quantitative reverse transcriptase PCR (scRT-PCR) to determine how chronic shock stress (CSS) affects electrophysiological and neurochemical properties of Type I, Type II, and Type III neurons in the BNSTALG. We report that CSS resulted in changes in the input resistance, time constant, action potential waveform, and firing rate of Type III but not Type I or II neurons. Additionally, only the Type III neurons exhibited an increase in Crf mRNA and a decrease in striatal-enriched protein tyrosine phosphatase (Ptpn5) mRNA after CSS. In contrast, only non-Type III cells showed a reduction in calcium-permeable AMPA receptor (CP-AMPAR) current and changes in mRNA expression of genes encoding AMPA receptor subunits after CSS. Importantly, none of the effects of CSS observed were seen in all cell types. Our results suggest that Type III neurons play a unique role in the BNSTALG circuit and represent a population of CRF neurons particularly sensitive to chronic stress.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Septal Nuclei/physiopathology , Stress, Psychological/physiopathology , Transcriptome , Animals , Male , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Septal Nuclei/metabolism , Stress, Psychological/metabolism
4.
Neurobiol Dis ; 124: 520-530, 2019 04.
Article in English | MEDLINE | ID: mdl-30593834

ABSTRACT

Adolescence represents a critical period of neurodevelopment, defined by structural and synaptic pruning within the prefrontal cortex. While characteristic of typical development, this structural instability may open a window of vulnerability to developing neuropsychiatric disorders, including depression. Thus, therapeutic interventions that support or expedite neural remodeling in adolescence may be advantageous. Here, we inhibited the neuronally-expressed cytoskeletal regulatory factor Rho-kinase (ROCK), focusing primarily on the clinically-viable ROCK inhibitor fasudil. ROCK inhibition had rapid antidepressant-like effects in adolescent mice, and its efficacy was comparable to ketamine and fluoxetine. It also modified levels of the antidepressant-related signaling factors, tropomyosin/tyrosine receptor kinase B and Akt, as well as the postsynaptic marker PSD-95, in the ventromedial prefrontal cortex (vmPFC). Meanwhile, adolescent-typical dendritic spine pruning on excitatory pyramidal neurons in the vmPFC was expedited. Further, vmPFC-specific shRNA-mediated reduction of ROCK2, the dominant ROCK isoform in the brain, had antidepressant-like consequences. We cautiously suggest that ROCK inhibitors may have therapeutic potential for adolescent-onset depression.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Antidepressive Agents/pharmacology , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Mice , Mice, Inbred C57BL , Prefrontal Cortex/enzymology , Protein Kinase Inhibitors/pharmacology
5.
Neurosci Biobehav Rev ; 90: 115-129, 2018 07.
Article in English | MEDLINE | ID: mdl-29660417

ABSTRACT

Confusion endures as to the exact role of the amygdala in relation to autism. To help resolve this we turned to the NIMH's Research Domain Criteria (RDoC) which provides a classification schema that identifies different categories of behaviors that can turn pathologic in mental health disorders, e.g. autism. While RDoC incorporates all the known neurobiological substrates for each domain, this review will focus primarily on the amygdala. We first consider the amygdala from an anatomical, historical, and developmental perspective. Next, we examine the different domains and constructs of RDoC that the amygdala is involved in: Negative Valence Systems, Positive Valence Systems, Cognitive Systems, Social Processes, and Arousal and Regulatory Systems. Then the evidence for a dysfunctional amygdala in autism is presented with a focus on alterations in development, prenatal valproic acid exposure as a model for ASD, and changes in the oxytocin system therein. Finally, a synthesis of RDoC, the amygdala, and autism is offered, emphasizing the task of disambiguation and suggestions for future research.


Subject(s)
Amygdala/physiopathology , Autistic Disorder/metabolism , Autistic Disorder/physiopathology , Cognition/physiology , Oxytocin/metabolism , Humans , National Institute of Mental Health (U.S.) , Research , United States , Valproic Acid/metabolism
7.
Brain Struct Funct ; 223(4): 1731-1745, 2018 May.
Article in English | MEDLINE | ID: mdl-29204911

ABSTRACT

Previous studies have shown that exposure to stressful events can enhance fear memory and anxiety-like behavior as well as increase synaptic plasticity in the rat basolateral amygdala (BLA). We have evidence that repeated unpredictable shock stress (USS) elicits a long-lasting increase in anxiety-like behavior in rats, but the cellular mechanisms mediating this response remain unclear. Evidence from recent morphological studies suggests that alterations in the dendritic arbor or spine density of BLA principal neurons may underlie stress-induced anxiety behavior. Recently, we have shown that the induction of long-term potentiation (LTP) in BLA principal neurons is dependent on activation of postsynaptic D1 dopamine receptors and the subsequent activation of the cyclic adenosine 5'-monophosphate (cAMP)-protein kinase A (PKA) signaling cascade. Here, we have used in vitro whole-cell patch-clamp recording from BLA principal neurons to investigate the long-term consequences of USS on their morphological properties and synaptic plasticity. We provided evidence that the enhanced anxiety-like behavior in response to USS was not associated with any significant change in the morphological properties of BLA principal neurons, but was associated with a changed frequency dependence of synaptic plasticity, lowered LTP induction threshold, and reduced expression of phosphodiesterase type 4 enzymes (PDE4s). Furthermore, pharmacological inhibition of PDE4 activity with rolipram mimics the effects of chronic stress on LTP induction threshold and baseline startle. Our results provide the first evidence that stress both enhances anxiety-like behavior and facilitates synaptic plasticity in the amygdala through a common mechanism of PDE4-mediated disinhibition of cAMP-PKA signaling.


Subject(s)
Basolateral Nuclear Complex/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Stress, Psychological/pathology , Acoustic Stimulation/adverse effects , Animals , Anxiety/etiology , Basolateral Nuclear Complex/physiopathology , Benzazepines/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Disease Models, Animal , Dopamine Antagonists/pharmacology , Down-Regulation/drug effects , Electric Stimulation , In Vitro Techniques , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Phosphodiesterase 4 Inhibitors/pharmacology , Psychoacoustics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reflex, Acoustic/drug effects , Reflex, Acoustic/physiology , Rolipram/pharmacology , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/etiology
8.
Neuropsychopharmacology ; 43(2): 373-383, 2018 01.
Article in English | MEDLINE | ID: mdl-28664926

ABSTRACT

The orbitofrontal cortex (OFC) is thought to link stimuli and actions with anticipated outcomes in order to sustain flexible behavior in an ever-changing environment. How it retains these associations to guide future behavior is less well-defined. Here we focused on one subregion of this heterogeneous structure, the ventrolateral OFC (VLO). CaMKII-driven inhibitory Gi-coupled designer receptors exclusively activated by designer drugs (DREADDs) were infused and subsequently activated by their ligand Clozapine-N-oxide (CNO) in conjunction with fear extinction training (a form of aversive conditioning) and response-outcome conditioning (a form of appetitive conditioning). Gi-DREADD-mediated inactivation of the VLO during extinction conditioning interfered with fear extinction memory, resulting in sustained freezing when mice were later tested drug-free. Similarly, Gi-DREADD-mediated inactivation in conjunction with response-outcome conditioning caused a later decay in goal-directed responding-that is, mice were unable to select actions based on the likelihood that they would be rewarded in a sustainable manner. By contrast, inhibitory Gi-DREADDs in the basolateral amygdala (BLA) impaired the acquisition of both conditioned fear extinction and response-outcome conditioning, as expected based on prior studies using other inactivation techniques. Meanwhile, DREADD-mediated inhibition of the dorsolateral striatum enhanced response-outcome conditioning, also in line with prior reports. Together, our findings suggest that learning-related neuroplasticity in the VLO may be necessary for memory retention in both appetitive and aversive domains.


Subject(s)
Basolateral Nuclear Complex/physiology , Behavior, Animal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Conditioning, Classical/physiology , Corpus Striatum/physiology , Designer Drugs/metabolism , Extinction, Psychological/physiology , Fear/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Receptors, G-Protein-Coupled , Retention, Psychology/physiology , Animals , Basolateral Nuclear Complex/drug effects , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Corpus Striatum/drug effects , Designer Drugs/administration & dosage , Extinction, Psychological/drug effects , Fear/drug effects , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Retention, Psychology/drug effects
9.
Neuropharmacology ; 126: 224-232, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28899729

ABSTRACT

The basolateral amygdala (BLA) is a key site for crossmodal association of sensory stimuli and an important relay in the neural circuitry of emotion. Indeed, the BLA receives substantial glutamatergic inputs from multiple brain regions including the prefrontal cortex and thalamic nuclei. Modulation of glutamatergic transmission in the BLA regulates stress- and anxiety-related behaviors. Serotonin (5-HT) also plays an important role in regulating stress-related behavior through activation of both pre- and postsynaptic 5-HT receptors. Multiple 5-HT receptors are expressed in the BLA, where 5-HT has been reported to modulate glutamatergic transmission. However, the 5-HT receptor subtype mediating this effect is not yet clear. The aim of this study was to use patch-clamp recordings from BLA neurons in an ex vivo slice preparation to examine 1) the effect of 5-HT on extrinsic sensory inputs, and 2) to determine if any pathway specificity exists in 5-HT regulation of glutamatergic transmission. Two independent input pathways into the BLA were stimulated: the external capsule to mimic cortical input, and the internal capsule to mimic thalamic input. Bath application of 5-HT reversibly reduced the amplitude of evoked excitatory postsynaptic currents (eEPSCs) induced by stimulation of both pathways. The decrease was associated with an increase in the paired-pulse ratio and coefficient of variation of eEPSC amplitude, suggesting 5-HT acts presynaptically. Moreover, the effect of 5-HT in both pathways was mimicked by the selective 5-HT1B receptor agonist CP93129, but not by the 5-HT1A receptor agonist 8-OH DPAT. Similarly the effect of exogenous 5-HT was blocked by the 5-HT1B receptor antagonist GR55562, but not affected by the 5-HT1A receptor antagonist WAY 100635 or the 5-HT2 receptor antagonists pirenperone and MDL 100907. Together these data suggest 5-HT gates cortical and thalamic glutamatergic inputs into the BLA by activating presynaptic 5-HT1B receptors.


Subject(s)
Basolateral Nuclear Complex/physiology , Cerebral Cortex/physiology , Glutamic Acid/physiology , Neurons/physiology , Serotonin/physiology , Thalamus/physiology , Animals , Benzamides/administration & dosage , Excitatory Postsynaptic Potentials , External Capsule/physiology , Internal Capsule/physiology , Male , Neural Pathways/physiology , Pyridines/administration & dosage , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B/physiology , Serotonin 5-HT1 Receptor Antagonists
10.
Mol Autism ; 8: 42, 2017.
Article in English | MEDLINE | ID: mdl-28775827

ABSTRACT

BACKGROUND: The amygdala controls socioemotional behavior and has consistently been implicated in the etiology of autism spectrum disorder (ASD). Precocious amygdala development is commonly reported in ASD youth with the degree of overgrowth positively correlated to the severity of ASD symptoms. Prenatal exposure to VPA leads to an ASD phenotype in both humans and rats and has become a commonly used tool to model the complexity of ASD symptoms in the laboratory. Here, we examined abnormalities in gene expression in the amygdala and socioemotional behavior across development in the valproic acid (VPA) rat model of ASD. METHODS: Rat dams received oral gavage of VPA (500 mg/kg) or saline daily between E11 and 13. Socioemotional behavior was tracked across development in both sexes. RNA sequencing and proteomics were performed on amygdala samples from male rats across development. RESULTS: Effects of VPA on time spent in social proximity and anxiety-like behavior were sex dependent, with social abnormalities presenting in males and heightened anxiety in females. Across time VPA stunted developmental and immune, but enhanced cellular death and disorder, pathways in the amygdala relative to saline controls. At postnatal day 10, gene pathways involved in nervous system and cellular development displayed predicted activations in prenatally exposed VPA amygdala samples. By juvenile age, however, transcriptomic and proteomic pathways displayed reductions in cellular growth and neural development. Alterations in immune pathways, calcium signaling, Rho GTPases, and protein kinase A signaling were also observed. CONCLUSIONS: As behavioral, developmental, and genomic alterations are similar to those reported in ASD, these results lend support to prenatal exposure to VPA as a useful tool for understanding how developmental insults to molecular pathways in the amygdala give rise to ASD-related syndromes.


Subject(s)
Amygdala , Autism Spectrum Disorder , Behavior, Animal/drug effects , Emotions/drug effects , Prenatal Exposure Delayed Effects , Social Behavior , Transcriptome/drug effects , Valproic Acid/adverse effects , Amygdala/metabolism , Amygdala/pathology , Amygdala/physiopathology , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Female , Humans , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Valproic Acid/pharmacology
11.
Nature ; 546(7657): 297-301, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28562592

ABSTRACT

Adult pair bonding involves dramatic changes in the perception and valuation of another individual. One key change is that partners come to reliably activate the brain's reward system, although the precise neural mechanisms by which partners become rewarding during sociosexual interactions leading to a bond remain unclear. Here we show, using a prairie vole (Microtus ochrogaster) model of social bonding, how a functional circuit from the medial prefrontal cortex to nucleus accumbens is dynamically modulated to enhance females' affiliative behaviour towards a partner. Individual variation in the strength of this functional connectivity, particularly after the first mating encounter, predicts how quickly animals begin affiliative huddling with their partner. Rhythmically activating this circuit in a social context without mating biases later preference towards a partner, indicating that this circuit's activity is not just correlated with how quickly animals become affiliative but causally accelerates it. These results provide the first dynamic view of corticostriatal activity during bond formation, revealing how social interactions can recruit brain reward systems to drive changes in affiliative behaviour.


Subject(s)
Arvicolinae/physiology , Arvicolinae/psychology , Nucleus Accumbens/physiology , Pair Bond , Prefrontal Cortex/physiology , Reward , Social Behavior , Animals , Female , Male , Mating Preference, Animal/physiology , Nucleus Accumbens/cytology , Prefrontal Cortex/cytology , Time Factors
12.
J Comp Neurol ; 525(9): 2235-2248, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28295315

ABSTRACT

The anterolateral group of the bed nucleus of the stria terminalis (BNSTALG ) is a critical modulator of a variety of rodent and primate behaviors spanning anxiety behavior and drug addiction. Three distinct neuronal cell types have been previously defined in the rat BNSTALG based on differences in the voltage-response to hyperpolarizing and depolarizing current injection. Differences in genetic expression profile between these three cell types suggest electrophysiological cell type may be an indicator for functional differences in the circuit of the rat BNSTALG . Although the behavioral role of the BNST is conserved across species, it is unknown if the same electrophysiological cell types exist in the BNSTALG of the mouse and nonhuman primate. Here, we used whole-cell patch clamp electrophysiology and neuronal reconstructions of biocytin-filled neurons to compare and contrast the electrophysiological and morphological properties of neurons in the BNSTALG from the mouse, rat, and rhesus macaque. We provide evidence that the BNSTALG of all three species contains neurons that match the three defined cell types found in the rat; however, there are intriguing differences in the relative frequency of these cell types as well as electrophysiological and morphological properties of the BNSTALG neurons across species. This study suggests that the overall landscape of the BNSTALG in the primate and mouse may be similar to that of the rat in some aspects but perhaps significantly different in others.


Subject(s)
Action Potentials/physiology , Neurons/classification , Neurons/physiology , Septal Nuclei/cytology , Analysis of Variance , Animals , Image Processing, Computer-Assisted , In Vitro Techniques , Lysine/analogs & derivatives , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Species Specificity
13.
Biol Psychiatry ; 81(4): 366-377, 2017 02 15.
Article in English | MEDLINE | ID: mdl-26786312

ABSTRACT

BACKGROUND: Distinguishing between actions that are more likely or less likely to be rewarded is a critical aspect of goal-directed decision making. However, neuroanatomic and molecular mechanisms are not fully understood. METHODS: We used anterograde tracing, viral-mediated gene silencing, functional disconnection strategies, pharmacologic rescue, and designer receptors exclusively activated by designer drugs (DREADDs) to determine the anatomic and functional connectivity between the orbitofrontal cortex (OFC) and the amygdala in mice. In particular, we knocked down brain-derived neurotrophic factor (Bdnf) bilaterally in the OFC or generated an OFC-amygdala "disconnection" by pairing unilateral OFC Bdnf knockdown with lesions of the contralateral amygdala. We characterized decision-making strategies using a task in which mice selected actions based on the likelihood that they would be reinforced. Additionally, we assessed the effects of DREADD-mediated OFC inhibition on the consolidation of action-outcome conditioning. RESULTS: As in other species, the OFC projects to the basolateral amygdala and dorsal striatum in mice. Bilateral Bdnf knockdown within the ventrolateral OFC and unilateral Bdnf knockdown accompanied by lesions of the contralateral amygdala impede goal-directed response selection, implicating BDNF-expressing OFC projection neurons in selecting actions based on their consequences. The tyrosine receptor kinase B agonist 7,8-dihydroxyflavone rescues action selection and increases dendritic spine density on excitatory neurons in the OFC. Rho-kinase inhibition also rescues goal-directed response strategies, linking neural remodeling with outcome-based decision making. Finally, DREADD-mediated OFC inhibition weakens new action-outcome memory. CONCLUSIONS: Activity-dependent and BDNF-dependent neuroplasticity within the OFC coordinate outcome-based decision making through interactions with the amygdala. These interactions break reward-seeking habits, a putative factor in multiple psychopathologies.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Decision Making/physiology , Goals , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Conditioning, Operant/physiology , Corpus Striatum/cytology , Corpus Striatum/physiology , Extinction, Psychological/physiology , Habits , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Receptor, trkB/physiology
14.
Nat Commun ; 7: 13149, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27767183

ABSTRACT

Molecular characterization of neuron populations, particularly those controlling threat responses, is essential for understanding the cellular basis of behaviour and identifying pharmacological agents acting selectively on fear-controlling circuitry. Here we demonstrate a comprehensive workflow for identification of pharmacologically tractable markers of behaviourally characterized cell populations. Thy1-eNpHR-, Thy1-Cre- and Thy1-eYFP-labelled neurons of the BLA consistently act as fear inhibiting or 'Fear-Off' neurons during behaviour. We use cell-type-specific optogenetics and chemogenetics (DREADDs) to modulate activity in this population during behaviour to block or enhance fear extinction. Dissociated Thy1-eYFP neurons are isolated using FACS. RNA sequencing identifies genes strongly upregulated in RNA of this population, including Ntsr2, Dkk3, Rspo2 and Wnt7a. Pharmacological manipulation of neurotensin receptor 2 confirms behavioural effects observed in optogenetic and chemogenetic experiments. These experiments identify and validate Ntsr2-expressing neurons within the BLA, as a putative 'Fear-Off' population.


Subject(s)
Basolateral Nuclear Complex/metabolism , Extinction, Psychological/physiology , Fear/physiology , Memory/physiology , Thy-1 Antigens/genetics , Adaptor Proteins, Signal Transducing , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/drug effects , Clozapine/analogs & derivatives , Clozapine/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Gene Expression Regulation , Genes, Reporter , Injections, Intraventricular , Integrases/genetics , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Memory/drug effects , Mice , Oligopeptides/pharmacology , Optogenetics/methods , Piperidines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sequence Analysis, RNA , Stereotaxic Techniques , Thrombospondins/genetics , Thrombospondins/metabolism , Thy-1 Antigens/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
15.
Brain Struct Funct ; 221(2): 839-54, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25381464

ABSTRACT

The basolateral nucleus of the amygdala (BLA) assigns emotional valence to sensory stimuli, and many amygdala-dependent behaviors undergo marked development during postnatal life. We recently showed principal neurons in the rat BLA undergo dramatic changes to their electrophysiological properties during the first postnatal month, but no study to date has thoroughly characterized changes to morphology or gene expression that may underlie the functional development of this neuronal population. We addressed this knowledge gap with reconstructions of biocytin-filled principal neurons in the rat BLA at postnatal days 7 (P7), 14, 21, 28, and 60. BLA principal neurons underwent a number of morphological changes, including a twofold increase in soma volume from P7 to P21. Dendritic arbors expanded significantly during the first postnatal month and achieved a mature distribution around P28, in terms of total dendritic length and distance from soma. The number of primary dendrites and branch points were consistent with age, but branch points were found farther from the soma in older animals. Dendrites of BLA principal neurons at P7 had few spines, and spine density increased nearly fivefold by P21. Given the concurrent increase in dendritic material, P60 neurons had approximately 17 times as many total spines as P7 neurons. Together, these developmental transitions in BLA principal neuron morphology help explain a number of concomitant electrophysiological changes during a critical period in amygdala development.


Subject(s)
Amygdala/physiology , Basolateral Nuclear Complex/physiology , Dendrites/physiology , Amygdala/metabolism , Animals , Basolateral Nuclear Complex/metabolism , Dendrites/metabolism , Male , Neurogenesis , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley
16.
Psychoneuroendocrinology ; 64: 66-78, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26615473

ABSTRACT

Loss of a partner can have severe effects on mental health. Here we explore the neural mechanisms underlying increased passive stress-coping, indicative of depressive-like behavior, following the loss of the female partner in the monogamous male prairie vole. We demonstrate that corticotropin-releasing factor receptor 2 (CRFR2) in the nucleus accumbens shell mediates social loss-induced passive coping. Further, we show that partner loss compromises the oxytocin system through multiple mechanisms. Finally, we provide evidence for an interaction of the CRFR2 and oxytocin systems in mediating the emotional consequences of partner loss. Our results suggest that chronic activation of CRFR2 and suppression of striatal oxytocin signaling following partner loss result in an aversive emotional state that may share underlying mechanisms with bereavement. We propose that the suppression of oxytocin signaling is likely adaptive during short separations to encourage reunion with the partner and may have evolved to maintain long-term partnerships. Additionally, therapeutic strategies targeting these systems should be considered for treatment of social loss-mediated depression.


Subject(s)
Adaptation, Psychological , Arvicolinae/physiology , Death , Nucleus Accumbens/physiology , Oxytocin/physiology , Pair Bond , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Autoradiography , Bacterial Proteins , Corpus Striatum/physiology , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/pharmacology , Female , Infusions, Intraventricular , Luminescent Proteins , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microinjections , Neurons/physiology , Nucleus Accumbens/drug effects , Oxytocin/administration & dosage , Oxytocin/pharmacology , Paraventricular Hypothalamic Nucleus/physiology , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Radioimmunoassay , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Urocortins/administration & dosage , Urocortins/pharmacology , Vasotocin/administration & dosage , Vasotocin/analogs & derivatives , Vasotocin/pharmacology
17.
Neuropsychopharmacology ; 41(1): 103-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26096838

ABSTRACT

The anterior bed nucleus of the stria terminalis (BNST) has been recognized as a critical structure in regulating trait anxiety, contextual fear memory, and appetitive behavior, and is known to be sensitive to stress manipulations. As one of the most complex structures in the central nervous system, the intrinsic circuitry of the BNST is largely unknown; however, recent technological developments have allowed researchers to begin to untangle the internal connections of the nucleus. This research has revealed the possibility of two opposing circuits, one anxiolytic and one anxiogenic, within the BNST, the relative strength of which determines the behavioral outcome. The balance of these pathways is critical in maintaining a normal physiological and behavioral state; however, stress and drugs of abuse can differentially affect the opposing circuitry within the nucleus to shift the balance to a pathological state. In this review, we will examine how stress interacts with the neuromodulators, corticotropin-releasing factor, norepinephrine, dopamine, and serotonin to affect the circuitry of the BNST as well as how synaptic plasticity in the BNST is modulated by stress, resulting in long-lasting changes in the circuit and behavioral state.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Nerve Net/metabolism , Septal Nuclei/metabolism , Stress, Psychological/metabolism , Animals , Dopamine/metabolism , Humans , Nerve Net/pathology , Neuronal Plasticity/physiology , Norepinephrine/metabolism , Septal Nuclei/pathology , Serotonin/metabolism , Stress, Psychological/pathology
18.
Neuropharmacology ; 97: 251-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26032436

ABSTRACT

Depression during pregnancy has been linked to in utero stress and is associated with long-lasting symptoms in offspring, including anxiety, helplessness, attentional deficits, and social withdrawal. Depression is diagnosed in 10-20% of expectant mothers, but the impact of antidepressant treatment on offspring development is not well documented, particularly for females. Here, we used a prenatal stress model of maternal depression to test the hypothesis that in utero antidepressant treatment could mitigate the effects of prenatal stress. We also investigated the effects of prenatal stress and antidepressant treatment on gene expression related to GABAergic and serotonergic neurotransmission in the amygdala, which may underlie behavioral effects of prenatal stress. Nulliparous female rats were implanted with osmotic minipumps delivering clinically-relevant concentrations of escitalopram and mated. Pregnant dams were exposed to 12 days of mixed-modality stressors, and offspring were behaviorally assessed in adolescence (postnatal day 28) and adulthood (beyond day 90) to determine the extent of behavioral change. We found that in utero stress exposure, regardless of escitalopram treatment, increased anxiety-like behavior in adolescent females and profoundly influenced amygdala expression of the chloride transporters KCC2 and NKCC1, which regulate GABAergic function. In contrast, prenatal escitalopram exposure alone elevated amygdala expression of 5-HT1A receptors. In adulthood, anxiety-like behavior returned to baseline and gene expression effects in the amygdala abated, whereas deficits emerged in novel object recognition for rats exposed to stress during gestation. These findings suggest prenatal stress causes age-dependent deficits in anxiety-like behavior and amygdala function in female offspring, regardless of antidepressant exposure.


Subject(s)
Amygdala/physiopathology , Anxiety/physiopathology , Citalopram/pharmacology , Prenatal Exposure Delayed Effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/physiopathology , Amygdala/drug effects , Amygdala/growth & development , Animals , Anxiety/etiology , Corticosterone/blood , Disease Models, Animal , Estradiol/blood , Female , Gene Expression/drug effects , Memory Disorders/etiology , Memory Disorders/physiopathology , Pregnancy , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Solute Carrier Family 12, Member 2/metabolism , Stress, Psychological/drug therapy , Symporters/metabolism , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
19.
J Clin Invest ; 125(4): 1497-508, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751059

ABSTRACT

Synaptic plasticity is the ability of synapses to modulate the strength of neuronal connections; however, the molecular factors that regulate this feature are incompletely understood. Here, we demonstrated that mice lacking brain-specific angiogenesis inhibitor 1 (BAI1) have severe deficits in hippocampus-dependent spatial learning and memory that are accompanied by enhanced long-term potentiation (LTP), impaired long-term depression (LTD), and a thinning of the postsynaptic density (PSD) at hippocampal synapses. We showed that compared with WT animals, mice lacking Bai1 exhibit reduced protein levels of the canonical PSD component PSD-95 in the brain, which stems from protein destabilization. We determined that BAI1 prevents PSD-95 polyubiquitination and degradation through an interaction with murine double minute 2 (MDM2), the E3 ubiquitin ligase that regulates PSD-95 stability. Restoration of PSD-95 expression in hippocampal neurons in BAI1-deficient mice by viral gene therapy was sufficient to compensate for Bai1 loss and rescued deficits in synaptic plasticity. Together, our results reveal that interaction of BAI1 with MDM2 in the brain modulates PSD-95 levels and thereby regulates synaptic plasticity. Moreover, these results suggest that targeting this pathway has therapeutic potential for a variety of neurological disorders.


Subject(s)
Angiogenic Proteins/physiology , Guanylate Kinases/metabolism , Hippocampus/physiopathology , Learning Disabilities/genetics , Membrane Proteins/metabolism , Memory Disorders/genetics , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-mdm2/physiology , Spatial Learning/physiology , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Brain/blood supply , Disks Large Homolog 4 Protein , Guanylate Kinases/deficiency , Guanylate Kinases/genetics , HEK293 Cells , Hippocampus/pathology , Humans , Learning Curve , Learning Disabilities/physiopathology , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Neurons/ultrastructure , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Synaptic Transmission/physiology , Ubiquitination
20.
Neuropsychopharmacology ; 40(9): 2135-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25716930

ABSTRACT

Prenatal stress (PS) is a risk factor for neurodevelopmental disorders with diverse ages of onset and socioemotional symptoms. Some PS-linked disorders involve characteristic social deficits, such as autism spectrum disorders and schizophrenia, but PS also promotes anxiety disorders. We propose the diversity of symptoms following PS arises from perturbations to early brain development. To this end, we characterized the effects of PS on the developmental trajectory of physiology of the amygdala, a late-developing center for socioemotional control. We found that PS dampened socioemotional behavior and reduced amygdala neuron excitability in offspring during infancy (at postnatal days (P)10, 14, 17 and 21), preadolescence (day 28), and adulthood (day 60). PS offspring in infancy produced fewer isolation-induced vocalizations and in adulthood exhibited less anxiety-like behavior and deficits in social interaction. PS neurons had a more hyperpolarized resting membrane potential from infancy to adulthood and produced fewer action potentials. Moreover, adult amygdala neurons from PS animals expressed larger action potential afterhyperpolarizations and H-current relative to controls, further limiting excitability. Our results suggest that PS can suppress socioemotional behavior throughout development and produce age-specific alterations to amygdala physiology.


Subject(s)
Amygdala/pathology , Developmental Disabilities/etiology , Neurons/physiology , Prenatal Exposure Delayed Effects , Social Behavior Disorders/etiology , Stress, Psychological/complications , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Female , In Vitro Techniques , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Sprague-Dawley , Vocalization, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...