Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
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
2.
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
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.
Development ; 147(6)2020 03 30.
Article in English | MEDLINE | ID: mdl-32094113

ABSTRACT

Noradrenaline belongs to the monoamine system and is involved in cognition and emotional behaviors. Phox2a and Phox2b play essential but non-redundant roles during development of the locus coeruleus (LC), the main noradrenergic (NA) neuron center in the mammalian brain. The ubiquitin E3 ligase Rnf220 and its cofactor Zc4h2 participate in ventral neural tube patterning by modulating Shh/Gli signaling, and ZC4H2 mutation is associated with intellectual disability, although the mechanisms for this remain poorly understood. Here, we report that Zc4h2 and Rnf220 are required for the development of central NA neurons in the mouse brain. Both Zc4h2 and Rnf220 are expressed in developing LC-NA neurons. Although properly initiated at E10.5, the expression of genes associated with LC-NA neurons is not maintained at the later embryonic stages in mice with a deficiency of either Rnf220 or Zc4h2 In addition, we show that the Rnf220/Zc4h2 complex monoubiquitylates Phox2a/Phox2b, a process required for the full transcriptional activity of Phox2a/Phox2b. Our work reveals a role for Rnf220/Zc4h2 in regulating LC-NA neuron development, and this finding may be helpful for understanding the pathogenesis of ZC4H2 mutation-associated intellectual disability.


Subject(s)
Adrenergic Neurons/physiology , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Neurogenesis/physiology , Nuclear Proteins/physiology , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination/genetics , Adrenergic Neurons/metabolism , Animals , Cell Differentiation/genetics , Chick Embryo , Embryo, Mammalian , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Norepinephrine/metabolism
6.
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
7.
J Neurosci ; 40(39): 7464-7474, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32868458

ABSTRACT

The neuropeptide galanin has been implicated in stress-related neuropsychiatric disorders in humans and rodent models. While pharmacological treatments for these disorders are ineffective for many individuals, physical activity is beneficial for stress-related symptoms. Galanin is highly expressed in the noradrenergic system, particularly the locus coeruleus (LC), which is dysregulated in stress-related disorders and activated by exercise. Galanin expression is elevated in the LC by chronic exercise, and blockade of galanin transmission attenuates exercise-induced stress resilience. However, most research on this topic has been done in rats, so it is unclear whether the relationship between exercise and galanin is species specific. Moreover, use of intracerebroventricular (ICV) galanin receptor antagonists in prior studies precluded defining a causal role for LC-derived galanin specifically. Therefore, the goals of this study were twofold. First, we investigated whether physical activity (chronic wheel running) increases stress resilience and galanin expression in the LC of male and female mice. Next, we used transgenic mice that overexpress galanin in noradrenergic neurons (Gal OX) to determine how chronically elevated noradrenergic-derived galanin, alone, alters anxiogenic-like responses to stress. We found that three weeks of ad libitum access to a running wheel in their home cage increased galanin mRNA in the LC of mice, which was correlated with and conferred resilience to stress. The effects of exercise were phenocopied by galanin overexpression in noradrenergic neurons, and Gal OX mice were resistant to the anxiogenic effect of optogenetic LC activation. These findings support a role for chronically increased noradrenergic galanin in mediating resilience to stress.SIGNIFICANCE STATEMENT Understanding the neurobiological mechanisms underlying behavioral responses to stress is necessary to improve treatments for stress-related neuropsychiatric disorders. Increased physical activity is associated with stress resilience in humans, but the neurobiological mechanisms underlying this effect are not clear. Here, we investigate a potential causal mechanism of this effect driven by the neuropeptide galanin from the main noradrenergic nucleus, the locus coeruleus (LC). We show that chronic voluntary wheel running in mice increases stress resilience and increases galanin expression in the LC. Furthermore, we show that genetic overexpression of galanin in noradrenergic neurons causes resilience to a stressor and the anxiogenic effects of optogenetic LC activation. These findings support a role for chronically increased noradrenergic galanin in mediating resilience to stress.


Subject(s)
Adrenergic Neurons/metabolism , Galanin/metabolism , Stress, Psychological/metabolism , Adrenergic Neurons/physiology , Animals , Female , Galanin/genetics , Male , Mice , Mice, Inbred C57BL , Motor Activity , Stress, Psychological/genetics , Stress, Psychological/physiopathology
8.
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
9.
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
10.
Cereb Cortex ; 30(9): 4979-4994, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32390051

ABSTRACT

The two catecholamines, noradrenaline and dopamine, have been shown to play comparable roles in behavior. Both noradrenergic and dopaminergic neurons respond to cues predicting reward availability and novelty. However, even though both are thought to be involved in motivating actions, their roles in motivation have seldom been directly compared. We therefore examined the activity of putative noradrenergic neurons in the locus coeruleus and putative midbrain dopaminergic neurons in monkeys cued to perform effortful actions for rewards. The activity in both regions correlated with engagement with a presented option. By contrast, only noradrenaline neurons were also (i) predictive of engagement in a subsequent trial following a failure to engage and (ii) more strongly activated in nonrepeated trials, when cues indicated a new task condition. This suggests that while both catecholaminergic neurons are involved in promoting action, noradrenergic neurons are sensitive to task state changes, and their influence on behavior extends beyond the immediately rewarded action.


Subject(s)
Adrenergic Neurons/physiology , Dopaminergic Neurons/physiology , Locus Coeruleus/physiology , Mesencephalon/physiology , Motivation/physiology , Animals , Macaca mulatta , Male , Reward
11.
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
12.
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
13.
J Neurosci ; 39(42): 8239-8249, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619493

ABSTRACT

The locus coeruleus (LC) is a seemingly singular and compact neuromodulatory nucleus that is a prominent component of disparate theories of brain function due to its broad noradrenergic projections throughout the CNS. As a diffuse neuromodulatory system, noradrenaline affects learning and decision making, control of sleep and wakefulness, sensory salience including pain, and the physiology of correlated forebrain activity (ensembles and networks) and brain hemodynamic responses. However, our understanding of the LC is undergoing a dramatic shift due to the application of state-of-the-art methods that reveal a nucleus of many modules that provide targeted neuromodulation. Here, we review the evidence supporting a modular LC based on multiple levels of observation (developmental, genetic, molecular, anatomical, and neurophysiological). We suggest that the concept of the LC as a singular nucleus and, alongside it, the role of the LC in diverse theories of brain function must be reconsidered.


Subject(s)
Adrenergic Neurons/physiology , Executive Function/physiology , Locus Coeruleus/physiology , Neurons/physiology , Animals , Humans , Nerve Net/physiology , Neural Pathways/physiology , Pain/physiopathology , Sleep/physiology , Wakefulness/physiology
14.
FASEB J ; 33(6): 7252-7260, 2019 06.
Article in English | MEDLINE | ID: mdl-30860868

ABSTRACT

Electroencephalogram monitoring during propofol (PRO) anesthesia typically features low-frequency oscillations, which may be involved with thalamic reticular nucleus (TRN) modulation. TRN receives noradrenergic inputs from the locus coeruleus (LC). We hypothesized that specific noradrenergic connections in the TRN may contribute to the emergence from PRO anesthesia. Intranuclei norepinephrine (NE) injections (n = 10) and designer receptors exclusively activated by designer drugs (DREADDs) (n = 10) were used to investigate the role of noradrenergic inputs from the LC to the TRN during PRO anesthesia. Whole-cell recording in acute brain slice preparations was used to identify the type of adrenoceptor that regulates noradrenergic innervation in the TRN. An intracerebral injection of NE into the TRN delays arousal in mice recovering from PRO anesthesia (means ± sd; 486.6 ± 57.32 s for the NE injection group vs. 422.4 ± 48.19 s for the control group; P = 0.0143) and increases the cortical-δ (0.1-4 Hz, 25.4 ± 2.9 for the NE injection group vs. 21.0 ± 1.7 for the control group; P = 0.0094) oscillation. An intra-TRN injection of NE also decreased the EC50 of PRO-induced unconsciousness (57.05 ± 1.78 mg/kg for the NE injection group vs. 72.44 ± 3.23 mg/kg for the control group; P = 0.0096). Moreover, the activation of LC-noradrenergic nerve terminals in the TRN using DREADDs increased the recovery time [466.1 ± 44.57 s for the clozapine N-oxide (CNO) injection group vs. 426.1 ± 38.75 s for the control group; P = 0.0033], decreased the EC50 of PRO-induced unconsciousness (64.77 ± 3.40 mg/kg for the CNO injection group vs. 74.00 ± 2.08 mg/kg for the control group; P = 0.0081), and increased the cortical-δ oscillation during PRO anesthesia (23.29 ± 2.58 for the CNO injection group vs. 19.56 ± 1.9 for the control group; P = 0.0213). In addition, whole-cell recording revealed that NE augmented the inhibitory postsynaptic currents in the TRN neurons via the α1-adrenoceptor. Our data indicated that enhanced NE signaling at the noradrenergic terminals of the LC-TRN projection delays arousal from general anesthesia, which is likely mediated by the α1-adrenoceptor activation. Our findings open a door for further understanding of the functions of various LC targets in both anesthesia and arousal.-Zhang, Y., Fu, B., Liu, C., Yu, S., Luo, T., Zhang, L., Zhou, W., Yu, T. Activation of noradrenergic terminals in the reticular thalamus delays arousal from propofol anesthesia in mice.


Subject(s)
Adrenergic Neurons/physiology , Anesthesia, General , Arousal/physiology , Delayed Emergence from Anesthesia/physiopathology , Intralaminar Thalamic Nuclei/physiopathology , Nerve Endings/physiology , Receptors, Adrenergic, alpha-1/physiology , Adrenergic Neurons/drug effects , Anesthetics, Intravenous , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Designer Drugs/pharmacology , Electroencephalography , Genetic Vectors/administration & dosage , Intralaminar Thalamic Nuclei/drug effects , Mice , Mice, Inbred C57BL , Nerve Endings/drug effects , Norepinephrine/pharmacology , Patch-Clamp Techniques , Propofol , Random Allocation , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/physiology , Receptors, Adrenergic, alpha-1/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Reflex, Righting/drug effects , Single-Blind Method , Specific Pathogen-Free Organisms , Synaptic Potentials/drug effects , Synaptic Potentials/physiology
15.
Mol Psychiatry ; 24(5): 710-725, 2019 05.
Article in English | MEDLINE | ID: mdl-30214043

ABSTRACT

Noradrenergic signaling plays a well-established role in promoting the stress response. Here we identify a subpopulation of noradrenergic neurons, defined by developmental expression of Hoxb1, that has a unique role in modulating stress-related behavior. Using an intersectional chemogenetic strategy, in combination with behavioral and physiological analyses, we show that activation of Hoxb1-noradrenergic (Hoxb1-NE) neurons decreases anxiety-like behavior and promotes an active coping strategy in response to acute stressors. In addition, we use cerebral blood volume-weighted functional magnetic resonance imaging to show that chemoactivation of Hoxb1-NE neurons results in reduced activity in stress-related brain regions, including the bed nucleus of the stria terminalis, amygdala, and locus coeruleus. Thus, the actions of Hoxb1-NE neurons are distinct from the well-documented functions of the locus coeruleus in promoting the stress response, demonstrating that the noradrenergic system contains multiple functionally distinct subpopulations.


Subject(s)
Adrenergic Neurons/physiology , Homeodomain Proteins/genetics , Stress, Physiological/genetics , Adaptation, Psychological/physiology , Adrenergic Neurons/metabolism , Amygdala/metabolism , Animals , Anxiety/genetics , Anxiety/metabolism , Behavior, Animal/physiology , Brain/metabolism , Female , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism
16.
Neurobiol Dis ; 128: 9-18, 2019 08.
Article in English | MEDLINE | ID: mdl-30149181

ABSTRACT

The pedunculopontine nucleus (PPN) is part of the mesencephalic locomotor region (MLR) and has been involved in the control of gait, posture, locomotion, sleep, and arousal. It likely participates in some motor and non-motor symptoms of Parkinson's disease and is regularly proposed as a surgical target to ameliorate gait, posture and sleep disorders in Parkinsonian patients. The PPN overlaps with the monoaminergic systems including dopamine, serotonin and noradrenaline in the modulation of the above-mentioned functions. All these systems are involved in Parkinson's disease and the mechanism of the anti-Parkinsonian agents, mostly L-DOPA. This suggests that PPN interacts with monoaminergic neurons and vice versa. Some evidence indicates that the PPN sends cholinergic, glutamatergic and even gabaergic inputs to mesencephalic dopaminergic cells, with the data regarding serotonergic or noradrenergic cells being less well known. Similarly, the control exerted by the PPN on dopaminergic neurons, is multiple and complex, and more extensively explored than the other monoaminergic systems. The data on the influence of monoaminergic systems on PPN neuron activity are rather scarce. While there is evidence that the PPN influences the therapeutic response of L-DOPA, it is still difficult to discerne the reciprocal action of the PPN and monoaminergic systems in this action. Additional data are required to better understand the functional organization of monoaminergic inputs to the MLR including the PPN to get a clearer picture of their interaction.


Subject(s)
Adrenergic Neurons/physiology , Antiparkinson Agents/therapeutic use , Dopaminergic Neurons/physiology , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Pedunculopontine Tegmental Nucleus/metabolism , Serotonergic Neurons/physiology , Animals , Antiparkinson Agents/pharmacology , Humans , Levodopa/pharmacology , Parkinson Disease/metabolism , Pedunculopontine Tegmental Nucleus/drug effects
17.
J Biol Inorg Chem ; 24(8): 1179-1188, 2019 12.
Article in English | MEDLINE | ID: mdl-31691104

ABSTRACT

Copper (Cu) plays an essential role in the development and function of the brain. In humans, genetic disorders of Cu metabolism may cause either severe Cu deficiency (Menkes disease) or excessive Cu accumulation (Wilson disease) in the brain tissue. In either case, the loss of Cu homeostasis results in catecholamine misbalance, abnormal myelination of neurons, loss of normal brain architecture, and a spectrum of neurologic and/or psychiatric manifestations. Several metabolic processes have been identified as particularly sensitive to Cu dis-homeostasis. This review focuses on the role of Cu in noradrenergic neurons and summarizes the current knowledge of mechanisms that maintain Cu homeostasis in these cells. The impact of Cu misbalance on catecholamine metabolism and functioning of noradrenergic system is discussed.


Subject(s)
Adrenergic Neurons/physiology , Copper/physiology , Locus Coeruleus/physiology , Adrenergic Neurons/metabolism , Animals , Catecholamines/metabolism , Copper/metabolism , Homeostasis/physiology , Humans , Ion Transport/physiology , Locus Coeruleus/metabolism
18.
Alcohol Clin Exp Res ; 43(8): 1695-1701, 2019 08.
Article in English | MEDLINE | ID: mdl-31141179

ABSTRACT

BACKGROUND: Relapse is a critical barrier to effective long-term treatment of alcoholism, and stress is often cited as a key trigger to relapse. Numerous studies suggest that stress-induced reinstatement to drug-seeking behaviors is mediated by norepinephrine (NE) and corticotropin-releasing factor (CRF) signaling interactions in the bed nucleus of the stria terminalis (BNST), a brain region critical to many behavioral and physiologic responses to stressors. Here, we sought to directly examine the effects of NE on BNST CRF neuron activity and determine whether these effects may be modulated by chronic intermittent EtOH (CIE) exposure or a single restraint stress. METHODS: Adult male CRF-tomato reporter mice were treatment-naïve, or either exposed to CIE for 2 weeks or to a single 1-hour restraint stress. Effects of application of exogenous NE on BNST CRF neuron activity were assessed via whole-cell patch-clamp electrophysiological techniques. RESULTS: We found that NE depolarized BNST CRF neurons in naïve mice in a ß-adrenergic receptor (AR)-dependent mechanism. CRF neurons from CIE- or stress-exposed mice had significantly elevated basal resting membrane potential compared to naïve mice. Furthermore, CIE and stress individually disrupted the ability of NE to depolarize CRF neurons, suggesting that both stress and CIE utilize ß-AR signaling to modulate BNST CRF neurons. Neither stress nor CIE altered the ability of exogenous NE to inhibit evoked glutamatergic transmission onto BNST CRF neurons as shown in naïve mice, a mechanism previously shown to be α-AR-dependent. CONCLUSIONS: Altogether, these findings suggest that stress and CIE interact with ß-AR signaling to modulate BNST CRF neuron activity, potentially disrupting the α/ß-AR balance of BNST CRF neuronal excitability. Restoration of α/ß-AR balance may lead to novel therapies for the alleviation of many stress-related disorders.


Subject(s)
Adrenergic Neurons/physiology , Corticotropin-Releasing Hormone/physiology , Ethanol/adverse effects , Norepinephrine/pharmacology , Restraint, Physical/physiology , Septal Nuclei/physiology , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Gene Knock-In Techniques , Glutamic Acid/physiology , Kynurenic Acid/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Norepinephrine/antagonists & inhibitors , Picrotoxin/pharmacology , Propranolol/pharmacology , Septal Nuclei/drug effects , Substance Withdrawal Syndrome/physiopathology
19.
Proc Natl Acad Sci U S A ; 113(50): E8169-E8177, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911798

ABSTRACT

Optogenetic methods to modulate cells and signaling pathways via targeted expression and activation of light-sensitive proteins have greatly accelerated the process of mapping complex neural circuits and defining their roles in physiological and pathological contexts. Recently demonstrated technologies based on injectable, microscale inorganic light-emitting diodes (µ-ILEDs) with wireless control and power delivery strategies offer important functionality in such experiments, by eliminating the external tethers associated with traditional fiber optic approaches. Existing wireless µ-ILED embodiments allow, however, illumination only at a single targeted region of the brain with a single optical wavelength and over spatial ranges of operation that are constrained by the radio frequency power transmission hardware. Here we report stretchable, multiresonance antennas and battery-free schemes for multichannel wireless operation of independently addressable, multicolor µ-ILEDs with fully implantable, miniaturized platforms. This advance, as demonstrated through in vitro and in vivo studies using thin, mechanically soft systems that separately control as many as three different µ-ILEDs, relies on specially designed stretchable antennas in which parallel capacitive coupling circuits yield several independent, well-separated operating frequencies, as verified through experimental and modeling results. When used in combination with active motion-tracking antenna arrays, these devices enable multichannel optogenetic research on complex behavioral responses in groups of animals over large areas at low levels of radio frequency power (<1 W). Studies of the regions of the brain that are involved in sleep arousal (locus coeruleus) and preference/aversion (nucleus accumbens) demonstrate the unique capabilities of these technologies.


Subject(s)
Optogenetics/instrumentation , Prostheses and Implants , Adrenergic Neurons/physiology , Animals , Arousal/physiology , Behavior, Animal , Deep Brain Stimulation/instrumentation , Electromagnetic Phenomena , Equipment Design , Locus Coeruleus/anatomy & histology , Locus Coeruleus/physiology , Locus Coeruleus/surgery , Male , Mice , Models, Theoretical , Reward , Wireless Technology/instrumentation
20.
Bull Exp Biol Med ; 168(2): 295-299, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31782006

ABSTRACT

Adrenergic innervation in the tissue of the thyroid gland, blood vessels of the thyroid gland, cervical lymphatic vessel, and lymph nodes in rats with hypothyroidism was studied by using a specific histochemical fluorescent-microscopic method of visualization of catecholamines. The presence of adrenergic innervation in the blood and lymph vessels and nodes was demonstrated. In hypothyroidism, diffusion of norepinephrine from nerve fibers and varicose thickenings was observed in the wall of the upper and lower thyroid arteries and adjacent cervical lymphatic vessels and nodes.


Subject(s)
Adrenergic Fibers/physiology , Blood Vessels/innervation , Hypothyroidism/pathology , Lymph Nodes/innervation , Lymphatic Vessels/innervation , Thyroid Gland/innervation , Adrenergic Neurons/physiology , Animals , Catecholamines/chemistry , Catecholamines/metabolism , Fluorescent Dyes/chemistry , Male , Nervous System/anatomy & histology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL