Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
Add more filters










Publication year range
1.
Nature ; 626(7998): 347-356, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267576

ABSTRACT

To survive in a complex social group, one needs to know who to approach and, more importantly, who to avoid. In mice, a single defeat causes the losing mouse to stay away from the winner for weeks1. Here through a series of functional manipulation and recording experiments, we identify oxytocin neurons in the retrochiasmatic supraoptic nucleus (SOROXT) and oxytocin-receptor-expressing cells in the anterior subdivision of the ventromedial hypothalamus, ventrolateral part (aVMHvlOXTR) as a key circuit motif for defeat-induced social avoidance. Before defeat, aVMHvlOXTR cells minimally respond to aggressor cues. During defeat, aVMHvlOXTR cells are highly activated and, with the help of an exclusive oxytocin supply from the SOR, potentiate their responses to aggressor cues. After defeat, strong aggressor-induced aVMHvlOXTR cell activation drives the animal to avoid the aggressor and minimizes future defeat. Our study uncovers a neural process that supports rapid social learning caused by defeat and highlights the importance of the brain oxytocin system in social plasticity.


Subject(s)
Aggression , Avoidance Learning , Hypothalamus , Neural Pathways , Neurons , Oxytocin , Social Learning , Animals , Mice , Aggression/physiology , Avoidance Learning/physiology , Cues , Fear/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Neural Pathways/physiology , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Social Learning/physiology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism , Neuronal Plasticity
2.
Nature ; 599(7883): 131-135, 2021 11.
Article in English | MEDLINE | ID: mdl-34646010

ABSTRACT

Oestrogen depletion in rodents and humans leads to inactivity, fat accumulation and diabetes1,2, underscoring the conserved metabolic benefits of oestrogen that inevitably decrease with age. In rodents, the preovulatory surge in 17ß-oestradiol (E2) temporarily increases energy expenditure to coordinate increased physical activity with peak sexual receptivity. Here we report that a subset of oestrogen-sensitive neurons in the ventrolateral ventromedial hypothalamic nucleus (VMHvl)3-7 projects to arousal centres in the hippocampus and hindbrain, and enables oestrogen to rebalance energy allocation in female mice. Surges in E2 increase melanocortin-4 receptor (MC4R) signalling in these VMHvl neurons by directly recruiting oestrogen receptor-α (ERα) to the Mc4r gene. Sedentary behaviour and obesity in oestrogen-depleted female mice were reversed after chemogenetic stimulation of VMHvl neurons expressing both MC4R and ERα. Similarly, a long-term increase in physical activity is observed after CRISPR-mediated activation of this node. These data extend the effect of MC4R signalling - the most common cause of monogenic human obesity8 - beyond the regulation of food intake and rationalize reported sex differences in melanocortin signalling, including greater disease severity of MC4R insufficiency in women9. This hormone-dependent node illuminates the power of oestrogen during the reproductive cycle in motivating behaviour and maintaining an active lifestyle in women.


Subject(s)
Brain/physiology , Estrogens/metabolism , Physical Exertion/physiology , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction , Animals , CRISPR-Cas Systems , Energy Metabolism , Estrogen Receptor alpha/metabolism , Estrogens/deficiency , Female , Gene Editing , Hippocampus/metabolism , Male , Melanocortins/metabolism , Mice , Neurons/metabolism , Obesity/metabolism , Rhombencephalon/metabolism , Sedentary Behavior , Sex Characteristics , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology
3.
Bull Exp Biol Med ; 171(2): 251-253, 2021 May.
Article in English | MEDLINE | ID: mdl-34173105

ABSTRACT

Spike activity of neurons in the ventromedial nucleus (VMN) of the hypothalamus in adult (6-8 months) and aged (2 years) male rats was studied by the in vivo extracellular method using stereotaxic insertion of microelectrodes. In all animals, firing frequency of most VMN neurons increased in response to glucose administration. However, in aged rats, the mean baseline and glucose-induced spike frequencies of VMN neurons were lower than in adult animals. These results support the hypothesis that aging is associated with a decrease in the functional activity of hypothalamic neurons.


Subject(s)
Aging/psychology , Ventromedial Hypothalamic Nucleus/physiology , Action Potentials/drug effects , Action Potentials/physiology , Aging/drug effects , Animals , Cortical Excitability/drug effects , Electrophysiological Phenomena/drug effects , Glucose/pharmacology , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/physiology , Insulin/pharmacology , Male , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
4.
Nat Commun ; 12(1): 2517, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947849

ABSTRACT

Survival depends on a balance between seeking rewards and avoiding potential threats, but the neural circuits that regulate this motivational conflict remain largely unknown. Using an approach-food vs. avoid-predator threat conflict test in rats, we identified a subpopulation of neurons in the anterior portion of the paraventricular thalamic nucleus (aPVT) which express corticotrophin-releasing factor (CRF) and are preferentially recruited during conflict. Inactivation of aPVTCRF neurons during conflict biases animal's response toward food, whereas activation of these cells recapitulates the food-seeking suppression observed during conflict. aPVTCRF neurons project densely to the nucleus accumbens (NAc), and activity in this pathway reduces food seeking and increases avoidance. In addition, we identified the ventromedial hypothalamus (VMH) as a critical input to aPVTCRF neurons, and demonstrated that VMH-aPVT neurons mediate defensive behaviors exclusively during conflict. Together, our findings describe a hypothalamic-thalamostriatal circuit that suppresses reward-seeking behavior under the competing demands of avoiding threats.


Subject(s)
Avoidance Learning/physiology , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/physiology , Midline Thalamic Nuclei/metabolism , Nerve Net/physiology , Neurons/metabolism , Ventromedial Hypothalamic Nucleus/physiology , Animals , Behavior Rating Scale , Conflict, Psychological , Female , Hypothalamus/metabolism , Male , Midline Thalamic Nuclei/cytology , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/radiation effects , Neurons/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Nucleus Accumbens/radiation effects , Optogenetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Reward , Ventromedial Hypothalamic Nucleus/cytology
5.
Elife ; 102021 05 21.
Article in English | MEDLINE | ID: mdl-34018926

ABSTRACT

The ventromedial hypothalamic nucleus (VMH) controls diverse behaviors and physiologic functions, suggesting the existence of multiple VMH neural subtypes with distinct functions. Combing translating ribosome affinity purification with RNA-sequencing (TRAP-seq) data with single-nucleus RNA-sequencing (snRNA-seq) data, we identified 24 mouse VMH neuron clusters. Further analysis, including snRNA-seq data from macaque tissue, defined a more tractable VMH parceling scheme consisting of six major genetically and anatomically differentiated VMH neuron classes with good cross-species conservation. In addition to two major ventrolateral classes, we identified three distinct classes of dorsomedial VMH neurons. Consistent with previously suggested unique roles for leptin receptor (Lepr)-expressing VMH neurons, Lepr expression marked a single dorsomedial class. We also identified a class of glutamatergic VMH neurons that resides in the tuberal region, anterolateral to the neuroanatomical core of the VMH. This atlas of conserved VMH neuron populations provides an unbiased starting point for the analysis of VMH circuitry and function.


Subject(s)
Multigene Family , Neurons/physiology , Transcriptome , Ventromedial Hypothalamic Nucleus/physiology , Animals , Cluster Analysis , Databases, Genetic , Gene Expression Profiling , Genotype , Glutamic Acid/metabolism , Macaca mulatta , Mice, Transgenic , Neurons/metabolism , Phenotype , RNA-Seq , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Species Specificity , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
6.
Neuron ; 109(7): 1150-1167.e6, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33600763

ABSTRACT

The hypothalamus plays crucial roles in regulating endocrine, autonomic, and behavioral functions via its diverse nuclei and neuronal subtypes. The developmental mechanisms underlying ontogenetic establishment of different hypothalamic nuclei and generation of neuronal diversity remain largely unknown. Here, we show that combinatorial T-box 3 (TBX3), orthopedia homeobox (OTP), and distal-less homeobox (DLX) expression delineates all arcuate nucleus (Arc) neurons and defines four distinct subpopulations, whereas combinatorial NKX2.1/SF1 and OTP/DLX expression identifies ventromedial hypothalamus (VMH) and tuberal nucleus (TuN) neuronal subpopulations, respectively. Developmental analysis indicates that all four Arc subpopulations are mosaically and simultaneously generated from embryonic Arc progenitors, whereas glutamatergic VMH neurons and GABAergic TuN neurons are sequentially generated from common embryonic VMH progenitors. Moreover, clonal lineage-tracing analysis reveals that diverse lineages from multipotent radial glia progenitors orchestrate Arc and VMH-TuN establishment. Together, our study reveals cellular mechanisms underlying generation and organization of diverse neuronal subtypes and ontogenetic establishment of individual nuclei in the mammalian hypothalamus.


Subject(s)
Hypothalamus/cytology , Hypothalamus/growth & development , Neurons/physiology , Animals , Animals, Genetically Modified , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/embryology , Cell Lineage , Glutamic Acid/physiology , Homeodomain Proteins/metabolism , Hypothalamus/embryology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Stem Cells/physiology , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/embryology , Ventromedial Hypothalamic Nucleus/metabolism , gamma-Aminobutyric Acid/physiology
7.
Diabetes ; 69(11): 2253-2266, 2020 11.
Article in English | MEDLINE | ID: mdl-32839348

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMN) is involved in the counterregulatory response to hypoglycemia. VMN neurons activated by hypoglycemia (glucose-inhibited [GI] neurons) have been assumed to play a critical although untested role in this response. Here, we show that expression of a dominant negative form of AMPK or inactivation of AMPK α1 and α2 subunit genes in Sf1 neurons of the VMN selectively suppressed GI neuron activity. We found that Txn2, encoding a mitochondrial redox enzyme, was strongly downregulated in the absence of AMPK activity and that reexpression of Txn2 in Sf1 neurons restored GI neuron activity. In cell lines, Txn2 was required to limit glucopenia-induced reactive oxygen species production. In physiological studies, absence of GI neuron activity after AMPK suppression in the VMN had no impact on the counterregulatory hormone response to hypoglycemia or on feeding. Thus, AMPK is required for GI neuron activity by controlling the expression of the antioxidant enzyme Txn2. However, the glucose-sensing capacity of VMN GI neurons is not required for the normal counterregulatory response to hypoglycemia. Instead, it may represent a fail-safe system in case of impaired hypoglycemia sensing by peripherally located glucose detection systems that are connected to the VMN.


Subject(s)
Glucose/metabolism , Hypoglycemia/blood , Neurons/physiology , Thioredoxins/metabolism , Ventromedial Hypothalamic Nucleus/cytology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Blood Glucose , Cells, Cultured , Humans , Patch-Clamp Techniques , Thioredoxins/genetics
8.
J Exp Zool A Ecol Integr Physiol ; 333(8): 550-560, 2020 10.
Article in English | MEDLINE | ID: mdl-32798281

ABSTRACT

The developing brain is highly sensitive to the hormonal milieu, with gonadal steroid hormones involved in neurogenesis, neural survival, and brain organization. Limited available evidence suggests that endocrine-disrupting chemicals (EDCs) may perturb these developmental processes. In this study, we tested the hypothesis that prenatal exposure to a mixture of polychlorinated biphenyls (PCBs), Aroclor 1221, would disrupt the normal timing of neurogenesis in two hypothalamic regions: the ventromedial nucleus (VMN) and the preoptic area (POA). These regions were selected because of their important roles in the control of sociosexual behaviors that are perturbed in adulthood by prenatal EDC exposure. Pregnant Sprague-Dawley rats were exposed to PCBs from Embryonic Day 8 (E8) to E18, encompassing the period of neurogenesis of all hypothalamic neurons. To determine the birth dates of neurons, bromo-2-deoxy-5-uridine (BrdU) was administered to dams on E12, E14, or E16. On the day after birth, male and female pups were perfused, brains immunolabeled for BrdU, and numbers of cells counted. In the VMN, exposure to PCBs significantly advanced the timing of neurogenesis compared to vehicle-treated pups, without changing the total number of BrdU+ cells. In the POA, PCBs did not change the timing of neurogenesis nor the total number of cells born. This is the first study to show that PCBs can shift the timing of neurogenesis in the hypothalamus, specifically in the VMN but not the POA. This result has implications for functions controlled by the VMN, especially sociosexual behaviors, as well as for sexual selection more generally.


Subject(s)
Endocrine Disruptors/pharmacology , Hypothalamus/drug effects , Neurogenesis/drug effects , Animals , Aroclors/pharmacology , Female , Fetus/drug effects , Neurons/drug effects , Polychlorinated Biphenyls/pharmacology , Pregnancy , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley , Sexual Behavior/drug effects , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
9.
Proc Natl Acad Sci U S A ; 117(32): 19566-19577, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719118

ABSTRACT

The ventromedial hypothalamus (VMH) plays chief roles regulating energy and glucose homeostasis and is sexually dimorphic. We discovered that expression of metabotropic glutamate receptor subtype 5 (mGluR5) in the VMH is regulated by caloric status in normal mice and reduced in brain-derived neurotrophic factor (BDNF) mutants, which are severely obese and have diminished glucose balance control. These findings led us to investigate whether mGluR5 might act downstream of BDNF to critically regulate VMH neuronal activity and metabolic function. We found that mGluR5 depletion in VMH SF1 neurons did not affect energy balance regulation. However, it significantly impaired insulin sensitivity, glycemic control, lipid metabolism, and sympathetic output in females but not in males. These sex-specific deficits are linked to reductions in intrinsic excitability and firing rate of SF1 neurons. Abnormal excitatory and inhibitory synapse assembly and elevated expression of the GABAergic synthetic enzyme GAD67 also cooperate to decrease and potentiate the synaptic excitatory and inhibitory tone onto mutant SF1 neurons, respectively. Notably, these alterations arise from disrupted functional interactions of mGluR5 with estrogen receptors that switch the normally positive effects of estrogen on SF1 neuronal activity and glucose balance control to paradoxical and detrimental. The collective data inform an essential central mechanism regulating metabolic function in females and underlying the protective effects of estrogen against metabolic disease.


Subject(s)
Blood Glucose/metabolism , Estrogens/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Ventromedial Hypothalamic Nucleus/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Energy Metabolism , Female , Glutamate Decarboxylase/metabolism , Homeostasis , Lipid Metabolism , Male , Mice , Mice, Mutant Strains , Nerve Net , Neural Inhibition , Neurons/metabolism , Neurons/physiology , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Estrogen/metabolism , Sex Factors , Signal Transduction , Steroidogenic Factor 1/metabolism , Sympathetic Nervous System/metabolism , Synaptic Transmission , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
10.
Nat Commun ; 11(1): 1729, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32265438

ABSTRACT

The TrkB receptor is critical for the control of energy balance, as mutations in its gene (NTRK2) lead to hyperphagia and severe obesity. The main neural substrate mediating the appetite-suppressing activity of TrkB, however, remains unknown. Here, we demonstrate that selective Ntrk2 deletion within paraventricular hypothalamus (PVH) leads to severe hyperphagic obesity. Furthermore, chemogenetic activation or inhibition of TrkB-expressing PVH (PVHTrkB) neurons suppresses or increases food intake, respectively. PVHTrkB neurons project to multiple brain regions, including ventromedial hypothalamus (VMH) and lateral parabrachial nucleus (LPBN). We find that PVHTrkB neurons projecting to LPBN are distinct from those to VMH, yet Ntrk2 deletion in PVH neurons projecting to either VMH or LPBN results in hyperphagia and obesity. Additionally, TrkB activation with BDNF increases firing of these PVH neurons. Therefore, TrkB signaling is a key regulator of a previously uncharacterized neuronal population within the PVH that impinges upon multiple circuits to govern appetite.


Subject(s)
Hyperphagia/metabolism , Membrane Glycoproteins/metabolism , Neurons/metabolism , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Appetite/genetics , Feeding Behavior/physiology , Female , Hyperphagia/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiopathology , Protein-Tyrosine Kinases/genetics , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
11.
Neuron ; 106(4): 637-648.e6, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32164875

ABSTRACT

Although the ventromedial hypothalamus ventrolateral area (VMHvl) is now well established as a critical locus for the generation of conspecific aggression, its role is complex, with neurons responding during multiple phases of social interactions with both males and females. It has been previously unclear how the brain uses this complex multidimensional signal and coordinates a discrete action: the attack. Here, we find a hypothalamic-midbrain circuit that represents hierarchically organized social signals during aggression. Optogenetic-assisted circuit mapping reveals a preferential projection from VMHvlvGlut2 to lPAGvGlut2 cells, and inactivation of downstream lPAGvGlut2 populations results in aggression-specific deficits. lPAG neurons are selective for attack action and exhibit short-latency, time-locked spiking relative to the activity of jaw muscles during biting. Last, we find that this projection conveys male-biased signals from the VMHvl to downstream lPAGvGlut2 neurons that are sensitive to features of ongoing activity, suggesting that action selectivity is generated by a combination of pre- and postsynaptic mechanisms.


Subject(s)
Aggression/physiology , Mesencephalon/physiology , Neural Pathways/physiology , Neurons/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Female , Male , Mesencephalon/cytology , Mice , Neural Pathways/cytology , Neurons/cytology , Ventromedial Hypothalamic Nucleus/cytology
12.
Brain Res Bull ; 157: 41-50, 2020 04.
Article in English | MEDLINE | ID: mdl-31981674

ABSTRACT

Mechanisms that underlie metabolic sensor acclimation to recurring insulin-induced hypoglycemia (RIIH) are unclear. Norepinephrine (NE) regulates ventromedial hypothalamic nucleus (VMN) gluco-stimulatory nitric oxide (NO) and gluco-inhibitory γ-aminobutryic acid (GABA) neuron signaling. Current research addressed the hypothesis that during RIIH, NE suppresses 5'-AMP-activated protein kinase (AMPK) reactivity in both populations and impedes counter-regulation. The brain is postulated to utilize non-glucose substrates, e.g. amino acids glutamine (Gln), glutamate (Glu), and aspartate (Asp), to produce energy during hypoglycemia. A correlated aim investigated whether NE controls pyruvate recycling pathway marker protein (glutaminase, GLT; malic enzyme, ME-1) expression in either metabolic-sensory cell population. Male rats were injected subcutaneously with vehicle or insulin on days 1-3, then pretreated on day 4 by intracerebroventricular delivery of the alpha1-adrenergic receptor (α1-AR) reverse-agonist prazocin (PRZ) or vehicle before final insulin therapy. PRZ prevented acute hypoglycemic augmentation of AMPK activation in each cell group. Antecedent hypoglycemic repression of sensor activity was reversed by PRZ in GABA neurons. During RIIH, nitrergic neurons exhibited α1-AR - dependent up-regulated GLT and α2-AR profiles, while GABA cells showed down-regulated α1-AR. LC-ESI-MS analysis documented a decline in VMN Glu, Gln, and Asp concentrations during acute hypoglycemia, and habituation of the former two profiles to RIIH. PRZ attenuated glucagon and corticosterone secretion during acute hypoglycemia, but reversed decrements in output of both hormones during RIIH. Results implicate adjustments in impact of α1-AR signaling in repressed VMN metabolic-sensory AMPK activation and counter-regulatory dysfunction during RIIH. Antecedent hypoglycemia may up-regulate NO neuron energy yield via α1-AR - mediated up-regulated pyruvate recycling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hypoglycemia/metabolism , Insulin/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Hypoglycemia/physiopathology , Hypoglycemic Agents/pharmacology , Male , Norepinephrine/metabolism , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/cytology
13.
J Mol Neurosci ; 70(5): 647-658, 2020 May.
Article in English | MEDLINE | ID: mdl-31925707

ABSTRACT

Brain glycogen is a vital energy source during metabolic imbalance. Metabolic sensory neurons in the ventromedial hypothalamic nucleus (VMN) shape glucose counter-regulation. Insulin-induced hypoglycemic (IIH) male rats were infused icv with the glycogen breakdown inhibitor CP-316,819 (CP) to investigate whether glycogen-derived fuel controls basal and/or hypoglycemic patterns of VMN gluco-regulatory neuron energy stability and transmitter signaling. CP caused dose-dependent amplification of basal VMN glycogen content and either mobilization (low dose) or augmentation (high dose) of this depot during IIH. Drug treatment also prevented hypoglycemic diminution of tissue glucose in multiple structures. Low CP dose caused IIH-reversible augmentation of AMPK activity and glutamate decarboxylase (GAD) protein levels in laser-microdissected VMN GABA neurons, while the higher dose abolished hypoglycemic adjustments in these profiles. VMN steroidogenic factor-1 (SF-1) neurons exhibited suppressed (low CP dose) or unchanged (high CP dose) basal SF-1 expression and AMPK refractoriness of hypoglycemia at each dose. CP caused dose-proportionate augmentation of neuronal nitric oxide synthase protein and enhancement (low dose) or diminution (high dose) of this profile during IIH; AMPK activity in these cells was decreased in high dose-pretreated IIH rats. CP exerted dose-dependent effects on basal and hypoglycemic patterns of glucagon, but not corticosterone secretion. Results verify that VMN GABA, SF-1, and nitrergic neurons are metabolic sensory in function and infer that these populations may screen unique aspects of neurometabolic instability. Correlation of VMN glycogen augmentation with attenuated hypoglycemic VMN gluco-regulatory neuron AMPK activity implies that expansion of this fuel reservoir preserves cellular energy stability during this metabolic threat.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen/metabolism , Indoles/pharmacology , Neurotransmitter Agents/metabolism , Phenylbutyrates/pharmacology , Protein Kinases/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Enzyme Inhibitors/administration & dosage , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Glycogen Phosphorylase/antagonists & inhibitors , Indoles/administration & dosage , Infusions, Intraventricular , Male , Phenylbutyrates/administration & dosage , Rats , Rats, Sprague-Dawley , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
14.
PLoS Comput Biol ; 15(6): e1007092, 2019 06.
Article in English | MEDLINE | ID: mdl-31158265

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMN) has an important role in diverse behaviours. The common involvement in these of sex steroids, nutritionally-related signals, and emotional inputs from other brain areas, suggests that, at any given time, its output is in one of a discrete number of possible states corresponding to discrete motivational drives. Here we explored how networks of VMN neurons might generate such a decision-making architecture. We began with minimalist assumptions about the intrinsic properties of VMN neurons inferred from electrophysiological recordings of these neurons in rats in vivo, using an integrate-and-fire based model modified to simulate activity-dependent post-spike changes in neuronal excitability. We used a genetic algorithm based method to fit model parameters to the statistical features of spike patterning in each cell. The spike patterns in both recorded cells and model cells were assessed by analysis of interspike interval distributions and of the index of dispersion of firing rate over different binwidths. Simpler patterned cells could be closely matched by single neuron models incorporating a hyperpolarising afterpotential and either a slow afterhyperpolarisation or a depolarising afterpotential, but many others could not. We then constructed network models with the challenge of explaining the more complex patterns. We assumed that neurons of a given type (with heterogeneity introduced by independently random patterns of external input) were mutually interconnected at random by excitatory synaptic connections (with a variable delay and a random chance of failure). Simple network models of one or two cell types were able to explain the more complex patterns. We then explored the information processing features of such networks that might be relevant for a decision-making network. We concluded that rhythm generation (in the slow theta range) and bistability arise as emergent properties of networks of heterogeneous VMN neurons.


Subject(s)
Decision Making/physiology , Models, Neurological , Ventromedial Hypothalamic Nucleus , Algorithms , Animals , Computational Biology , Male , Neurons/cytology , Neurons/physiology , Rats , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology
15.
Proc Natl Acad Sci U S A ; 116(15): 7503-7512, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30898882

ABSTRACT

Type 1 estrogen receptor-expressing neurons in the ventrolateral subdivision of the ventromedial hypothalamus (VMHvlEsr1) play a causal role in the control of social behaviors, including aggression. Here we use six different viral-genetic tracing methods to systematically map the connectional architecture of VMHvlEsr1 neurons. These data reveal a high level of input convergence and output divergence ("fan-in/fan-out") from and to over 30 distinct brain regions, with a high degree (∼90%) of bidirectionality, including both direct as well as indirect feedback. Unbiased collateralization mapping experiments indicate that VMHvlEsr1 neurons project to multiple targets. However, we identify two anatomically distinct subpopulations with anterior vs. posterior biases in their collateralization targets. Nevertheless, these two subpopulations receive indistinguishable inputs. These studies suggest an overall system architecture in which an anatomically feed-forward sensory-to-motor processing stream is integrated with a dense, highly recurrent central processing circuit. This architecture differs from the "brain-inspired," hierarchical feed-forward circuits used in certain types of artificial intelligence networks.


Subject(s)
Behavior, Animal/physiology , Nerve Net/physiology , Neurons/metabolism , Social Behavior , Ventromedial Hypothalamic Nucleus/physiology , Animals , Brain Mapping , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Mice , Mice, Transgenic , Nerve Net/cytology , Neurons/cytology , Ventromedial Hypothalamic Nucleus/cytology
16.
Nat Neurosci ; 22(4): 565-575, 2019 04.
Article in English | MEDLINE | ID: mdl-30804529

ABSTRACT

Avoidance of innate threats is often in conflict with motivations to engage in exploratory approach behavior. The neural pathways that mediate this approach-avoidance conflict are not well resolved. Here we isolated a population of dopamine D1 receptor (D1R)-expressing neurons within the posteroventral region of the medial amygdala (MeApv) in mice that are activated either during approach or during avoidance of an innate threat stimulus. Distinct subpopulations of MeApv-D1R neurons differentially innervate the ventromedial hypothalamus and bed nucleus of the stria terminalis, and these projections have opposing effects on investigation or avoidance of threatening stimuli. These projections are potently modulated through opposite actions of D1R signaling that bias approach behavior. These data demonstrate divergent pathways in the MeApv that can be differentially weighted toward exploration or evasion of threats.


Subject(s)
Avoidance Learning/physiology , Choice Behavior/physiology , Conflict, Psychological , Corticomedial Nuclear Complex/physiology , Neurons/physiology , Receptors, Dopamine D1/physiology , Animals , Corticomedial Nuclear Complex/cytology , Fear/physiology , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/physiology , Septal Nuclei/cytology , Ventromedial Hypothalamic Nucleus/cytology
17.
Mol Cell Neurosci ; 95: 51-58, 2019 03.
Article in English | MEDLINE | ID: mdl-30660767

ABSTRACT

The ventromedial hypothalamic nucleus (VMN) is a critical component of the neural circuitry that regulates glucostasis. Astrocyte glycogen is a vital reserve of glucose and its oxidizable metabolite L-lactate. In hypoglycemic female rats, estradiol-dependent augmentation of VMN glycogen phosphorylase (GP) protein requires hindbrain catecholamine input. Research here investigated the premise that norepinephrine (NE) regulation of VMN astrocyte metabolism shapes local glucoregulatory neurotransmitter signaling in this sex. Estradiol-implanted ovariectomized rats were pretreated by intra-VMN administration of the monocarboxylate transporter inhibitor alpha-cyano-4-hydroxy-cinnamic acid (4CIN) or vehicle before NE delivery to that site. NE caused 4CIN-reversible reduction or augmentation of VMN glycogen synthase and phosphorylase expression. 4CIN prevented NE stimulation of gluco-inhibitory (glutamate decarboxylase65/67) and suppression of gluco-stimulatory (neuronal nitric oxide synthase) neuron marker proteins. These outcomes imply that effects of noradrenergic stimulation of VMN astrocyte glycogen depletion on glucoregulatory transmitter signaling may be mediated, in part, by glycogen-derived substrate fuel provision. NE control of astrocyte glycogen metabolism may involve down-regulated adrenoreceptor (AR), e.g. alpha1 and alpha2, alongside amplified beta1 AR and estrogen receptor-beta signaling. Noradrenergic hypoglycemia was refractory to 4CIN, implying that additional NE-sensitive VMN glucoregulatory neurochemicals may be insensitive to monocarboxylate uptake. Augmentation of circulating free fatty acids by combinatory NE and 4CIN, but not NE alone implies that acute hypoglycemia induced here is an insufficient stimulus for mobilization of these fuels, but is adequate when paired with diminished brain monocarboxylate fuel availability.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Norepinephrine/pharmacology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Astrocytes/metabolism , Coumaric Acids/pharmacology , Enzyme Inhibitors/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/deficiency , Fatty Acids/metabolism , Female , Glycogen Synthase/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/metabolism , Ventromedial Hypothalamic Nucleus/cytology
18.
Nature ; 564(7735): 213-218, 2018 12.
Article in English | MEDLINE | ID: mdl-30518859

ABSTRACT

Although the hippocampus is known to be important for declarative memory, it is less clear how hippocampal output regulates motivated behaviours, such as social aggression. Here we report that pyramidal neurons in the CA2 region of the hippocampus, which are important for social memory, promote social aggression in mice. This action depends on output from CA2 to the lateral septum, which is selectively enhanced immediately before an attack. Activation of the lateral septum by CA2 recruits a circuit that disinhibits a subnucleus of the ventromedial hypothalamus that is known to trigger attack. The social hormone arginine vasopressin enhances social aggression by acting on arginine vasopressin 1b receptors on CA2 presynaptic terminals in the lateral septum to facilitate excitatory synaptic transmission. In this manner, release of arginine vasopressin in the lateral septum, driven by an animal's internal state, may serve as a modulatory control that determines whether CA2 activity leads to declarative memory of a social encounter and/or promotes motivated social aggression.


Subject(s)
Aggression/physiology , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Neural Inhibition , Neural Pathways/physiology , Septal Nuclei/cytology , Septal Nuclei/physiology , Social Behavior , Animals , Arginine Vasopressin/metabolism , Clozapine/analogs & derivatives , Clozapine/pharmacology , Excitatory Postsynaptic Potentials , Female , Male , Memory/physiology , Mice , Mice, Inbred BALB C , Motivation , Presynaptic Terminals/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Pyramidal Cells/metabolism , Receptors, Vasopressin/metabolism , Synaptic Transmission , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology
19.
eNeuro ; 5(3)2018.
Article in English | MEDLINE | ID: mdl-29971248

ABSTRACT

The basomedial amygdala (BM) influences the ventromedial nucleus of the hypothalamus (VMH) through direct glutamatergic projections as well as indirectly, through the anterior part of the bed nucleus of the stria terminalis (BNSTa). However, BM and BNSTa axons end in a segregated fashion in VMH. BM projects to the core of VMH, where VMH's projection cells are located, whereas BNSTa projects to the shell of VMH, where GABAergic cells that inhibit core neurons are concentrated. However, the consequences of this dual regulation of VMH by BM and BNSTa are unknown. To study this question, we recorded the responses of VMH's shell and core neurons to the optogenetic activation of BM or BNSTa inputs in transgenic mice that selectively express Cre-recombinase in glutamatergic or GABAergic neurons. Glutamatergic BM inputs fired most core neurons but elicited no response in GABAergic shell neurons. Following BM infusions of AAV-EF1α-DIO-hChR2-mCherry in Vgat-ires-Cre-Ai6 mice, no anterograde labeling was observed in the VMH, suggesting that GABAergic BM neurons do not project to the VMH. In contrast, BNSTa sent mostly GABAergic projections that inhibited both shell and core neurons. However, BNSTa-evoked IPSPs had a higher amplitude in shell neurons. Since we also found that activation of GABAergic shell neurons causes an inhibition of core neurons, these results suggest that depending on the firing rate of shell neurons, BNSTa inputs could elicit a net inhibition or disinhibition of core neurons. Thus, the dual regulation of VMH by BM and BNSTa imparts flexibility to this regulator of defensive and social behaviors.


Subject(s)
Corticomedial Nuclear Complex/physiology , Neurons/physiology , Septal Nuclei/physiology , Ventromedial Hypothalamic Nucleus/physiology , Action Potentials , Animals , Corticomedial Nuclear Complex/cytology , Female , GABAergic Neurons/cytology , GABAergic Neurons/physiology , Male , Mice , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Optogenetics , Septal Nuclei/cytology , Ventromedial Hypothalamic Nucleus/cytology
20.
Elife ; 72018 06 15.
Article in English | MEDLINE | ID: mdl-29905528

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP, Adcyap1) is a neuromodulator implicated in anxiety, metabolism and reproductive behavior. PACAP global knockout mice have decreased fertility and PACAP modulates LH release. However, its source and role at the hypothalamic level remain unknown. We demonstrate that PACAP-expressing neurons of the ventral premamillary nucleus of the hypothalamus (PMVPACAP) project to, and make direct contact with, kisspeptin neurons in the arcuate and AVPV/PeN nuclei and a subset of these neurons respond to PACAP exposure. Targeted deletion of PACAP from the PMV through stereotaxic virally mediated cre- injection or genetic cross to LepR-i-cre mice with Adcyap1fl/fl mice led to delayed puberty onset and impaired reproductive function in female, but not male, mice. We propose a new role for PACAP-expressing neurons in the PMV in the relay of nutritional state information to regulate GnRH release by modulating the activity of kisspeptin neurons, thereby regulating reproduction in female mice.


Subject(s)
Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Reproduction/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/cytology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Reproduction/genetics , Sex Factors , Sexual Maturation/genetics , Ventromedial Hypothalamic Nucleus/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...