Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
eNeuro ; 11(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39160074

ABSTRACT

Adaptive behavior relies on efficient cognitive control. The anterior cingulate cortex (ACC) is a key node within the executive prefrontal network. The reciprocal connectivity between the locus ceruleus (LC) and ACC is thought to support behavioral reorganization triggered by the detection of an unexpected change. We transduced LC neurons with either excitatory or inhibitory chemogenetic receptors in adult male rats and trained rats on a spatial task. Subsequently, we altered LC activity and confronted rats with an unexpected change of reward locations. In a new spatial context, rats with decreased noradrenaline (NA) in the ACC entered unbaited maze arms more persistently which was indicative of perseveration. In contrast, the suppression of the global NA transmission reduced perseveration. Neither chemogenetic manipulation nor inactivation of the ACC by muscimol affected the rate of learning, possibly due to partial virus transduction of the LC neurons and/or the compensatory engagement of other prefrontal regions. Importantly, we observed behavioral deficits in rats with LC damage caused by virus injection. The latter finding highlights the importance of careful histological assessment of virus-transduced brain tissue as inadvertent damage of the targeted cell population due to virus neurotoxicity or other factors might cause unwanted side effects. Although the specific role of ACC in the flexibility of spatial behavior has not been convincingly demonstrated, our results support the beneficial role of noradrenergic transmission for an optimal function of the ACC. Overall, our findings suggest the LC exerts the projection-specific modulation of neural circuits mediating the flexibility of spatial behavior.


Subject(s)
Gyrus Cinguli , Locus Coeruleus , Norepinephrine , Spatial Behavior , Animals , Male , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Norepinephrine/metabolism , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiology , Spatial Behavior/physiology , Spatial Behavior/drug effects , Rats , Muscimol/pharmacology , Maze Learning/physiology , Maze Learning/drug effects , Neural Pathways/drug effects , Neural Pathways/physiology , Adrenergic Neurons/drug effects , Adrenergic Neurons/physiology
2.
Metabolism ; 157: 155940, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38878857

ABSTRACT

BACKGROUND AND AIM: Although it is well established that hormones like glucagon stimulates gluconeogenesis via the PKA-mediated phosphorylation of CREB and dephosphorylation of the cAMP-regulated CREB coactivators CRTC2, the role of neural signals in the regulation of gluconeogenesis remains uncertain. METHODS AND RESULTS: Here, we characterize the noradrenergic bundle architecture in mouse liver; we show that the sympathoexcitation induced by acute cold exposure promotes hyperglycemia and upregulation of gluconeogenesis via triggering of the CREB/CRTC2 pathway. Following its induction by dephosphorylation, CRTC2 translocates to the nucleus and drives the transcription of key gluconeogenic genes. Rodents submitted to different models of sympathectomy or knockout of CRTC2 do not activate gluconeogenesis in response to cold. Norepinephrine directly acts in hepatocytes mainly through a Ca2+-dependent pathway that stimulates CREB/CRTC2, leading to activation of the gluconeogenic program. CONCLUSION: Our data demonstrate the importance of the CREB/CRTC2 pathway in mediating effects of hepatic sympathetic inputs on glucose homeostasis, providing new insights into the role of norepinephrine in health and disease.


Subject(s)
Cold Temperature , Cyclic AMP Response Element-Binding Protein , Gluconeogenesis , Liver , Norepinephrine , Transcription Factors , Animals , Gluconeogenesis/physiology , Liver/metabolism , Mice , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Norepinephrine/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Adrenergic Neurons/metabolism , Adrenergic Neurons/physiology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Hepatocytes/metabolism
3.
Eur J Neurosci ; 60(1): 3659-3676, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38872397

ABSTRACT

The locus coeruleus (LC) is the primary source of noradrenergic transmission in the mammalian central nervous system. This small pontine nucleus consists of a densely packed nuclear core-which contains the highest density of noradrenergic neurons-embedded within a heterogeneous surround of non-noradrenergic cells. This local heterogeneity, together with the small size of the LC, has made it particularly difficult to infer noradrenergic cell identity based on extracellular sampling of in vivo spiking activity. Moreover, the relatively high cell density, background activity and synchronicity of LC neurons have made spike identification and unit isolation notoriously challenging. In this study, we aimed at bridging these gaps by performing juxtacellular recordings from single identified neurons within the mouse LC complex. We found that noradrenergic neurons (identified by tyrosine hydroxylase, TH, expression; TH-positive) and intermingled putatively non-noradrenergic (TH-negative) cells displayed similar morphologies and responded to foot shock stimuli with excitatory responses; however, on average, TH-positive neurons exhibited more prominent foot shock responses and post-activation firing suppression. The two cell classes also displayed different spontaneous firing rates, spike waveforms and temporal spiking properties. A logistic regression classifier trained on spontaneous electrophysiological features could separate the two cell classes with 76% accuracy. Altogether, our results reveal in vivo electrophysiological correlates of TH-positive neurons, which can be useful for refining current approaches for the classification of LC unit activity.


Subject(s)
Action Potentials , Adrenergic Neurons , Locus Coeruleus , Locus Coeruleus/physiology , Locus Coeruleus/cytology , Animals , Mice , Male , Action Potentials/physiology , Adrenergic Neurons/physiology , Mice, Inbred C57BL , Neurons/physiology , Tyrosine 3-Monooxygenase/metabolism
4.
J Neurophysiol ; 132(1): 68-77, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38838298

ABSTRACT

The prepositus hypoglossi nucleus (PHN) and the interstitial nucleus of Cajal (INC) are involved in the control of horizontal and vertical gaze, respectively. A previous study showed that PHN neurons exhibit depolarized or hyperpolarized responses to noradrenaline (NA). However, the adrenoceptor types that participate in NA-induced responses and the effects of NA on INC neurons have not yet been investigated. Furthermore, the relationship between NA-induced responses and neuron types defined by neurotransmitter phenotypes has not been determined. In this study, we investigated NA-induced current responses in PHN and INC neurons and the relationships between these responses and neuron types using whole cell recordings in wild-type and transgenic rat brainstem slices. Local application of NA to the cell soma induced slow inward (SI) and slow outward (SO) currents that were mainly mediated by α1 and α2 adrenoceptors, respectively. These current responses were observed in both PHN and INC neurons, although the proportion of INC neurons that responded to NA was low. Analyses of the distributions of the current responses revealed that in the PHN, all fluorescently identified inhibitory neurons exhibited SI currents, whereas glutamatergic and cholinergic neurons exhibited both SI and SO currents. In the INC, glutamatergic and inhibitory neurons preferentially exhibited SI and SO currents, respectively. When the PHN and INC neurons were characterized by their firing pattern, we found that the proportions of the currents depended on their firing pattern. These results suggest that various modes of noradrenergic modulation in horizontal and vertical neural integrators are dependent on neuron type.NEW & NOTEWORTHY Noradrenergic modulation of oculomotor neural integrators involved in gaze control has not been elucidated. Here, we report that noradrenaline (NA)-induced slow inward (SI) and outward (SO) currents are mediated mainly by α1 and α2 adrenoceptors in neurons that participate in horizontal and vertical gaze control. The NA-induced current responses differed depending on the neurotransmitter phenotype and firing pattern. These results suggest various modes of noradrenergic modulation in horizontal and vertical integrator neurons.


Subject(s)
Norepinephrine , Animals , Norepinephrine/pharmacology , Rats , Male , Rats, Transgenic , Neurons/physiology , Neurons/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-1/physiology , Adrenergic Neurons/physiology , Adrenergic Neurons/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, alpha-2/physiology , Patch-Clamp Techniques , Brain Stem/physiology , Brain Stem/cytology , Brain Stem/drug effects , Cholinergic Neurons/physiology , Cholinergic Neurons/drug effects
5.
J Neurosci ; 44(29)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38744530

ABSTRACT

Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMS without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus ceruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector, which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.


Subject(s)
Locus Coeruleus , Rats, Sprague-Dawley , Receptor, Melatonin, MT1 , Sleep, REM , Animals , Male , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Locus Coeruleus/physiology , Rats , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/metabolism , Sleep, REM/physiology , Sleep, REM/drug effects , Norepinephrine/metabolism , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Adrenergic Neurons/physiology , Neurons/metabolism , Neurons/drug effects , Neurons/physiology
6.
Curr Biol ; 34(12): 2644-2656.e7, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38810638

ABSTRACT

An epidemic of sleep loss currently affects modern societies worldwide and is implicated in numerous physiological disorders, including pain sensitization, although few studies have explored the brain pathways affected by active sleep deprivation (ASD; e.g., due to recreation). Here, we describe a neural circuit responsible for pain sensitization in mice treated with 9-h non-stress ASD. Using a combination of advanced neuroscience methods, we found that ASD stimulates noradrenergic inputs from locus coeruleus (LCNA) to glutamatergic neurons of the hindlimb primary somatosensory cortex (S1HLGlu). Moreover, artificial inhibition of this LCNA→S1HLGlu pathway alleviates ASD-induced pain sensitization in mice, while chemogenetic activation of this pathway recapitulates the pain sensitization observed following ASD. Our study thus implicates activation of the LCNA→S1HLGlu pathway in ASD-induced pain sensitization, expanding our fundamental understanding of the multisystem interplay involved in pain processing.


Subject(s)
Locus Coeruleus , Pain , Sleep Deprivation , Somatosensory Cortex , Animals , Mice , Sleep Deprivation/physiopathology , Locus Coeruleus/metabolism , Locus Coeruleus/physiopathology , Pain/physiopathology , Somatosensory Cortex/physiopathology , Male , Norepinephrine/metabolism , Mice, Inbred C57BL , Adrenergic Neurons/metabolism , Adrenergic Neurons/physiology , Neurons/physiology , Neurons/metabolism , Neural Pathways/physiopathology
7.
Cell Stem Cell ; 30(9): 1166-1178.e8, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37597516

ABSTRACT

The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that ß-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.


Subject(s)
Adrenergic Neurons , Immunity, Innate , Intestinal Mucosa , Lymphocytes , Regeneration , Animals , Signal Transduction , Adrenergic Neurons/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Mice , Interleukin-22
8.
Elife ; 122023 02 03.
Article in English | MEDLINE | ID: mdl-36734517

ABSTRACT

The locus coeruleus (LC) houses the vast majority of noradrenergic neurons in the brain and regulates many fundamental functions, including fight and flight response, attention control, and sleep/wake cycles. While efferent projections of the LC have been extensively investigated, little is known about its local circuit organization. Here, we performed large-scale multipatch recordings of noradrenergic neurons in adult mouse LC to profile their morpho-electric properties while simultaneously examining their interactions. LC noradrenergic neurons are diverse and could be classified into two major morpho-electric types. While fast excitatory synaptic transmission among LC noradrenergic neurons was not observed in our preparation, these mature LC neurons connected via gap junction at a rate similar to their early developmental stage and comparable to other brain regions. Most electrical connections form between dendrites and are restricted to narrowly spaced pairs or small clusters of neurons of the same type. In addition, more than two electrically coupled cell pairs were often identified across a cohort of neurons from individual multicell recording sets that followed a chain-like organizational pattern. The assembly of LC noradrenergic neurons thus follows a spatial and cell-type-specific wiring principle that may be imposed by a unique chain-like rule.


Subject(s)
Adrenergic Neurons , Locus Coeruleus , Mice , Animals , Locus Coeruleus/physiology , Adrenergic Neurons/physiology , Synaptic Transmission , Attention
9.
Am J Physiol Regul Integr Comp Physiol ; 323(4): R512-R531, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35993562

ABSTRACT

In mammals, the pontine noradrenergic system influences nearly every aspect of central nervous system function. A subpopulation of pontine noradrenergic neurons, called A5, are thought to be important in the cardiovascular response to physical stressors, yet their function is poorly defined. We hypothesized that activation of A5 neurons drives a sympathetically mediated increase in blood pressure (BP). To test this hypothesis, we conducted a comprehensive assessment of the cardiovascular effects of chemogenetic stimulation of A5 neurons in male and female adult rats using intersectional genetic and anatomical targeting approaches. Chemogenetic stimulation of A5 neurons in freely behaving rats elevated BP by 15 mmHg and increased cardiac baroreflex sensitivity with a negligible effect on resting HR. Importantly, A5 stimulation had no detectable effect on locomotor activity, metabolic rate, or respiration. Under anesthesia, stimulation of A5 neurons produced a marked elevation in visceral sympathetic nerve activity (SNA) and no change in skeletal muscle SNA, showing that A5 neurons preferentially stimulate visceral SNA. Interestingly, projection mapping indicates that A5 neurons target sympathetic preganglionic neurons throughout the spinal cord and parasympathetic preganglionic neurons throughout in the brainstem, as well as the nucleus of the solitary tract, and ventrolateral medulla. Moreover, in situ hybridization and immunohistochemistry indicate that a subpopulation of A5 neurons coreleases glutamate and monoamines. Collectively, this study suggests A5 neurons are a central modulator of autonomic function with a potentially important role in sympathetically driven redistribution of blood flow from the visceral circulation to critical organs and skeletal muscle.


Subject(s)
Adrenergic Neurons , Adrenergic Neurons/physiology , Animals , Blood Pressure/physiology , Female , Glutamates/pharmacology , Male , Mammals , Pons/physiology , Rats , Sympathetic Nervous System/physiology
10.
Proc Natl Acad Sci U S A ; 119(18): e2116507119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35486692

ABSTRACT

The noradrenergic locus coeruleus (LC) is a controller of brain and behavioral states. Activating LC neurons en masse by electrical or optogenetic stimulation promotes a stereotypical "activated" cortical state of high-frequency oscillations. However, it has been recently reported that spontaneous activity of LC cell pairs has sparse yet structured time-averaged cross-correlations, which is unlike the highly synchronous neuronal activity evoked by stimulation. Therefore, LC population activity could consist of distinct multicell ensembles each with unique temporal evolution of activity. We used nonnegative matrix factorization (NMF) to analyze large populations of simultaneously recorded LC single units in the rat LC. NMF identified ensembles of spontaneously coactive LC neurons and their activation time courses. Since LC neurons selectively project to specific forebrain regions, we hypothesized that distinct ensembles activate during different cortical states. To test this hypothesis, we calculated band-limited power and spectrograms of local field potentials in cortical area 24a aligned to spontaneous activations of distinct LC ensembles. A diversity of state modulations occurred around activation of different LC ensembles, including a typical activated state with increased high-frequency power as well as other states including decreased high-frequency power. Thus­in contrast to the stereotypical activated brain state evoked by en masse LC stimulation­spontaneous activation of distinct LC ensembles is associated with a multitude of cortical states.


Subject(s)
Adrenergic Neurons , Locus Coeruleus , Adrenergic Neurons/physiology , Arousal/physiology , Locus Coeruleus/physiology , Norepinephrine , Optogenetics
11.
Brain Res ; 1777: 147754, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34929182

ABSTRACT

A long-standing observation is that the micturition reflex receives supraspinal descending control. Although one supraspinal nucleus (Barrington's nucleus) is identified as the pontine micturition center, it remains largely unknown whether and how other supraspinal tracts are involved in micturition control. Here, we focused on the role of lumbosacral projecting neurons located in the Locus Coeruleus (LC) in modulating micturition, since previous studies indicated that the LC is involved in controlling bladder contraction. First, by performing an AAV mediated retrograde labeling using a TH-iCre mouse line, we demonstrated specific targeting of LC noradrenergic neurons innervating the lumbosacral spinal cord with high efficiency. Next, by lumbosacral injection of a retro-AAV carrying Cre-dependent human diphtheria toxin receptors (DTR), we achieved specific ablation of LC NA+ neurons with lumbosacral projections upon the administration of diphtheria toxin. Our results showed that specific ablation of theseneurons led to overflow incontinence leaks and lower void efficiency. Mechanistically, by performing the urodynamics analysis, we showed that ablation of lumbosacral innervating NAneurons resulted in detrusor-sphincter dyssynergia. Taken together, our study provided novel insights into the underlying mechanisms of supraspinal control of micturition reflex and thus shed light on developing novel treatment to improve micturition control in patients with SCI or lower urinary tract symptoms.


Subject(s)
Adrenergic Neurons/physiology , Spinal Cord/physiology , Urinary Bladder/innervation , Urination/physiology , Animals , Locus Coeruleus/physiology , Mice , Pons/physiology , Reflex/physiology
12.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298984

ABSTRACT

While much of biomedical research since the middle of the twentieth century has focused on molecular pathways inside the cell, there is increasing evidence that extracellular signaling pathways are also critically important in health and disease. The neuromodulators norepinephrine (NE), serotonin (5-hydroxytryptamine, 5HT), dopamine (DA), acetylcholine (ACH), and melatonin (MT) are extracellular signaling molecules that are distributed throughout the brain and modulate many disease processes. The effects of these five neuromodulators on Alzheimer's disease (AD) are briefly examined in this paper, and it is hypothesized that each of the five molecules has a u-shaped (or Janus-faced) dose-response curve, wherein too little or too much signaling is pathological in AD and possibly other diseases. In particular it is suggested that NE is largely functionally opposed to 5HT, ACH, MT, and possibly DA in AD. In this scenario, physiological "balance" between the noradrenergic tone and that of the other three or four modulators is most healthy. If NE is largely functionally opposed to other prominent neuromodulators in AD, this may suggest novel combinations of pharmacological agents to counteract this disease. It is also suggested that the majority of cases of AD and possibly other diseases involve an excess of noradrenergic tone and a collective deficit of the other four modulators.


Subject(s)
Alzheimer Disease/physiopathology , Neurotransmitter Agents/antagonists & inhibitors , Norepinephrine/physiology , Synaptic Transmission/physiology , Adrenergic Agents/administration & dosage , Adrenergic Agents/therapeutic use , Adrenergic Neurons/physiology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Brain Chemistry , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Humans , Melatonin/therapeutic use , Mice , Models, Neurological , Neurotransmitter Agents/physiology , Nootropic Agents/pharmacology , Nootropic Agents/therapeutic use , Norepinephrine/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Rats , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Adrenergic, alpha-2/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/therapeutic use , Signal Transduction/drug effects , Synaptic Transmission/drug effects , tau Proteins/metabolism
13.
Neuron ; 109(13): 2116-2130.e6, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34081911

ABSTRACT

Dysregulation in contextual processing is believed to affect several forms of psychopathology, such as post-traumatic stress disorder (PTSD). The dentate gyrus (DG), a subregion of the hippocampus, is thought to be an important brain region for disambiguating new experiences from prior experiences. Noradrenergic (NE) neurons in the locus coeruleus (LC) are more tonically active during stressful events and send dense projections to the DG, yet an understanding of their function in DG-dependent contextual discrimination has not been established. Here, we isolate a key function of the LC-NE-DG circuit in contextual aversive generalization using selective manipulations and in vivo single-cell calcium imaging. We report that activation of LC-NE neurons and terminal activity results in contextual generalization. We found that these effects required ß-adrenergic-mediated modulation of hilar interneurons to ultimately promote aversive generalization, suggesting that disruption of noradrenergic tone may serve as an important avenue for treating stress-induced disorders.


Subject(s)
Adrenergic Neurons/physiology , Dentate Gyrus/physiology , Fear/physiology , Generalization, Psychological/physiology , Locus Coeruleus/physiology , Animals , Conditioning, Classical/physiology , Female , Male , Mice, Inbred C57BL
14.
Behav Brain Res ; 411: 113367, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34000338

ABSTRACT

Exposure to stress might influence pain sensitivity; however, little is known about whether post-traumatic stress disorder (PTSD)-like symptoms alter pain sensitivity and how it can happen. Male rats were exposed to the inescapable footshock paired with either social isolation or a control condition (not exposed to footshock but subjected to social isolation). After 7, 14, or 21 days, memory retention was evaluated. In the following three days, animals underwent the following tests: open-field, social interaction and formalin tests. Another group of animals were subjected to the object recognition test and to von Frey filaments. In other cohorts of animals, saline, fluoxetine, or desipramine were injected intrathecally and immunohistochemistry was performed to investigate whether PTSD-like symptoms alter the expression of c-Fos in serotonergic and noradrenergic neurons. Inescapable footshock induced the development of PTSD-like symptoms. Animals with PTSD-like symptoms showed an increase in the number of flinches in the formalin test and a reduction in mechanical threshold in the von Frey test at both retention intervals. The social interaction was negatively correlated with the nociceptive response in the formalin test. Fluoxetine or desipramine prevented the nociceptive response to chemical stimulus in the formalin test. In addition, in animals with PTSD-like symptoms, there was a reduction in c-Fos expression in serotonergic and noradrenergic neurons. Our results are important for the association of increased sensitivity to pain as one of the clinical manifestations that are present in the development of PTSD, and a possible treatment for increased pain sensitivity in male individuals with PTSD.


Subject(s)
Pain/physiopathology , Stress Disorders, Post-Traumatic/physiopathology , Adrenergic Neurons/metabolism , Adrenergic Neurons/physiology , Animals , Behavior, Animal , Fluoxetine/pharmacology , Male , Norepinephrine/metabolism , Pain/metabolism , Pain Management/psychology , Pain Threshold/drug effects , Rats , Rats, Wistar , Serotonergic Neurons/metabolism , Serotonergic Neurons/physiology , Social Behavior , Stress Disorders, Post-Traumatic/metabolism
15.
Pflugers Arch ; 473(6): 859-872, 2021 06.
Article in English | MEDLINE | ID: mdl-33855632

ABSTRACT

The pontine A5 noradrenergic group contributes to the maturation of the respiratory system before birth in rats. These neurons are connected to the neural network responsible for respiratory rhythmogenesis. In the present study, we investigated the participation of A5 noradrenergic neurons in neonates (P7-8 and P14-15) in the control of ventilation during hypoxia and hypercapnia in in vivo experiments using conjugated saporin anti-dopamine beta-hydroxylase (DßH-SAP) to specifically ablate noradrenergic neurons. Thus, DßH-SAP (420 ng/µL) or saporin (SAP, control) was injected into the A5 region of neonatal male Wistar rats. Hypoxia reduced respiratory variability in control animals; however, A5 lesion prevented this effect in P7-8 rats. Our data suggest that noradrenergic neurons of the A5 region in neonate rats do not participate in the control of ventilation under baseline and hypercapnic conditions, but exert an inhibitory modulation on breathing variability under hypoxic challenge in early life (P7-8).


Subject(s)
Adrenergic Neurons/metabolism , Brain Stem/cytology , Hypercapnia/physiopathology , Hypoxia/physiopathology , Respiration , Adrenergic Neurons/drug effects , Adrenergic Neurons/physiology , Animals , Animals, Newborn , Brain Stem/growth & development , Brain Stem/physiopathology , Dopamine beta-Hydroxylase/pharmacology , Male , Rats , Rats, Wistar , Saporins/pharmacology
16.
Bull Exp Biol Med ; 170(5): 585-589, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33792819

ABSTRACT

General anesthesia with Nembutal (40 mg/kg) dramatically decreased the power of all waves of HRV spectrum in rats, especially in LF and VLF frequency bands, but the HR and respiration rate were little changed. At this, individual spectral peaks in HF range were observed at the same frequencies (1.3-1.5 Hz), which are characteristic of the wakeful state. Preliminary stimulation of noradrenergic system with maprotiline (10 mg/kg) increased the power of HF waves and elevated the respiratory rate in narcotized rats in comparison with the control values, although it did not shift the spectral peak at 1.5 Hz in frequency axis. Preliminary stimulation of cholinergic system with galantamine (2 mg/kg) somewhat decreased the power of HF waves and respiratory rate in narcotized rats (in comparison with the control values); additionally, it shifted HF peak to 1.1-1.4 Hz. Activation of serotonergic system with 5-hydroxytryptophan (50 mg/kg) and fluoxetine (3 mg/kg) decreased the HR, the power of HF waves, and respiratory rate in narcotized rats. It also shifted the spectral peak of HF waves to 0.9-0.95 Hz. Preliminary stimulation of dopaminergic system with L-DOPA (20 ml/kg) and amantadine (20 ml/kg) increased the power of VLF waves in narcotized rats in comparison with the control values. Numerous peaks appeared in HF (1.1-1.2 Hz) and VLF frequency bands. Generally, preliminary stimulation of serotonergic or dopaminergic systems markedly affects the neural activity under following general anesthesia: first aggravates the effect of anesthesia on vital centers in CNS, whereas second weakens the effect of anesthesia at the suprasegmental level of neural control.


Subject(s)
Adrenergic Neurons/physiology , Heart Rate/physiology , Anesthesia , Animals , Male , Neurotransmitter Agents/metabolism , Rats , Receptors, Neurotransmitter/metabolism , Respiratory Rate/physiology
17.
Psychopharmacology (Berl) ; 238(7): 1765-1779, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33649970

ABSTRACT

RATIONALE: The flashing lights and sounds of modern casinos are alluring and may contribute to the addictive nature of gambling. Such cues can have a profound impact on the noradrenaline (NA) system, which could therefore be a viable therapeutic target for gambling disorder (GD). While there is substantial evidence to support the involvement of NA in the impulsive symptoms of GD, its function in mediating the "pro-addictive" impact of cues is less understood. OBJECTIVE: We wished to investigate the role of NA in our rodent assay of decision making and impulsivity, the cued rat gambling task (crGT). Given that sex differences are prominent in addiction disorders, and increasingly reported in the monoaminergic regulation of behaviour, we also prioritised evaluating noradrenergic drugs in both sexes. METHODS: Female and male rats were trained to stability on the crGT and then given intraperitoneal injections of the noradrenaline reuptake inhibitor atomoxetine, the α2A receptor agonist guanfacine, the beta receptor antagonist propranolol, and the α2 receptor antagonist yohimbine. RESULTS: Atomoxetine dose-dependently improved decision-making score. Guanfacine selectively enhanced decision making in risk-preferring males and optimal performing females. Propranolol and yohimbine did not influence decision making. Atomoxetine and guanfacine reduced premature responses, while yohimbine bi-phasically affected this index of motor impulsivity. CONCLUSIONS: These results support the hypothesis that NA is an important neuromodulator of the cue-induced deficits in decision making observed in laboratory-based gambling paradigms, and suggest that NAergic drugs like atomoxetine and guanfacine may be useful in treating GD.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Cues , Gambling/psychology , Impulsive Behavior/drug effects , Risk-Taking , Adrenergic Neurons/drug effects , Adrenergic Neurons/physiology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Atomoxetine Hydrochloride/pharmacology , Atomoxetine Hydrochloride/therapeutic use , Decision Making/drug effects , Decision Making/physiology , Dose-Response Relationship, Drug , Female , Gambling/drug therapy , Guanfacine/pharmacology , Guanfacine/therapeutic use , Impulsive Behavior/physiology , Male , Norepinephrine/pharmacology , Norepinephrine/therapeutic use , Rats , Rats, Long-Evans , Reaction Time/drug effects , Reaction Time/physiology
18.
Nat Neurosci ; 24(5): 646-657, 2021 05.
Article in English | MEDLINE | ID: mdl-33753944

ABSTRACT

Children with autism spectrum disorder often exhibit delays in achieving motor developmental milestones such as crawling, walking and speech articulation. However, little is known about the neural mechanisms underlying motor-related deficits. Here, we reveal that mice with a syntenic deletion of the chromosome 16p11.2, a common copy number variation associated with autism spectrum disorder, also exhibit delayed motor learning without showing gross motor deficits. Using in vivo two-photon imaging in awake mice, we find that layer 2/3 excitatory neurons in the motor cortex of adult male 16p11.2-deletion mice show abnormally high activity during the initial phase of learning, and the process of learning-induced spine reorganization is prolonged. Pharmacogenetic activation of locus coeruleus noradrenergic neurons was sufficient to rescue the circuit deficits and the delayed motor learning in these mice. Our results unveil an unanticipated role of noradrenergic neuromodulation in improving the delayed motor learning in 16p11.2-deletion male mice.


Subject(s)
Adrenergic Neurons/physiology , Autistic Disorder/physiopathology , Chromosome Deletion , Learning/physiology , Locus Coeruleus/physiopathology , Motor Skills/physiology , Animals , Autistic Disorder/genetics , Chromosomes, Mammalian , DNA Copy Number Variations , Disease Models, Animal , Microscopy, Fluorescence, Multiphoton
20.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R342-R361, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33296280

ABSTRACT

Previously, we identified a population of neurons in the hindbrain tegmentum, bordering the locus coeruleus (LC). We named this population the pre-locus coeruleus (pre-LC) because in rats its neurons lie immediately rostral to the LC. In mice, however, pre-LC and LC neurons intermingle, making them difficult to distinguish. Here, we use molecular markers and anterograde tracing to clarify the location and distribution of pre-LC neurons in mice, relative to rats. First, we colocalized the transcription factor FoxP2 with the activity marker Fos to identify pre-LC neurons in sodium-deprived rats and show their distribution relative to surrounding catecholaminergic and cholinergic neurons. Next, we used sodium depletion and chemogenetic activation of the aldosterone-sensitive HSD2 neurons in the nucleus of the solitary tract (NTS) to identify the homologous population of pre-LC neurons in mice, along with a related population in the central lateral parabrachial nucleus. Using Cre-reporter mice for Pdyn, we confirmed that most of these sodium-depletion-activated neurons are dynorphinergic. Finally, after confirming that these neurons receive excitatory input from the NTS and paraventricular hypothalamic nucleus, plus convergent input from the inhibitory AgRP neurons in the arcuate hypothalamic nucleus, we identify a major, direct input projection from the medial prefrontal cortex. This new information on the location, distribution, and input to pre-LC neurons provides a neuroanatomical foundation for cell-type-specific investigation of their properties and functions in mice. Pre-LC neurons likely integrate homeostatic information from the brainstem and hypothalamus with limbic, contextual information from the cerebral cortex to influence ingestive behavior.


Subject(s)
Brain/physiology , Neural Pathways/physiology , Neurons/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adrenergic Neurons/physiology , Animal Feed , Animals , Appetite Regulation , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Brain/cytology , Brain/metabolism , Cholinergic Neurons/physiology , Diet, Sodium-Restricted , Enkephalins/genetics , Feeding Behavior , Female , Locus Coeruleus/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/metabolism , Protein Precursors/genetics , Rats, Sprague-Dawley , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL