1
|
Luppi PH, Malcey J, Chancel A, Duval B, Cabrera S, Fort P. Neuronal network controlling REM sleep. J Sleep Res 2024:e14266. [PMID: 38972672 DOI: 10.1111/jsr.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024]
Abstract
Rapid eye movement sleep is a state characterized by concomitant occurrence of rapid eye movements, electroencephalographic activation and muscle atonia. In this review, we provide up to date knowledge on the neuronal network controlling its onset and maintenance. It is now accepted that muscle atonia during rapid eye movement sleep is due to activation of glutamatergic neurons localized in the pontine sublaterodorsal tegmental nucleus. These neurons directly project and excite glycinergic/γ-aminobutyric acid-ergic pre-motoneurons localized in the ventromedial medulla. The sublaterodorsal tegmental nucleus rapid eye movement-on neurons are inactivated during wakefulness and non-rapid eye movement by rapid eye movement-off γ-aminobutyric acid-ergic neurons localized in the ventrolateral periaqueductal grey and the adjacent dorsal deep mesencephalic reticular nucleus. Melanin-concentrating hormone and γ-aminobutyric acid-ergic rapid eye movement sleep-on neurons localized in the lateral hypothalamus would inhibit these rapid eye movement sleep-off neurons initiating the state. Finally, the activation of a few limbic cortical structures during rapid eye movement sleep by the claustrum and the supramammillary nucleus as well as that of the basolateral amygdala would be involved in the function(s) of rapid eye movement sleep. In summary, rapid eye movement sleep is generated by a brainstem generator controlled by forebrain structures involved in autonomic control.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Justin Malcey
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Amarine Chancel
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Blandine Duval
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
| | - Sébastien Cabrera
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Patrice Fort
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| |
Collapse
|
2
|
Luppi PH, Chancel A, Malcey J, Cabrera S, Fort P, Maciel RM. Which structure generates paradoxical (REM) sleep: The brainstem, the hypothalamus, the amygdala or the cortex? Sleep Med Rev 2024; 74:101907. [PMID: 38422648 DOI: 10.1016/j.smrv.2024.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 03/02/2024]
Abstract
Paradoxical or Rapid eye movement (REM) sleep (PS) is a state characterized by REMs, EEG activation and muscle atonia. In this review, we discuss the contribution of brainstem, hypothalamic, amygdalar and cortical structures in PS genesis. We propose that muscle atonia during PS is due to activation of glutamatergic neurons localized in the pontine sublaterodorsal tegmental nucleus (SLD) projecting to glycinergic/GABAergic pre-motoneurons localized in the ventro-medial medulla (vmM). The SLD PS-on neurons are inactivated during wakefulness and slow-wave sleep by PS-off GABAergic neurons localized in the ventrolateral periaqueductal gray (vPAG) and the adjacent deep mesencephalic reticular nucleus. Melanin concentrating hormone (MCH) and GABAergic PS-on neurons localized in the posterior hypothalamus would inhibit these PS-off neurons to initiate the state. Finally, the activation of a few limbic cortical structures during PS by the claustrum and the supramammillary nucleus as well as that of the basolateral amygdala would also contribute to PS expression. Accumulating evidence indicates that the activation of these limbic structures plays a role in memory consolidation and would communicate to the PS-generating structures the need for PS to process memory. In summary, PS generation is controlled by structures distributed from the cortex to the medullary level of the brain.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France.
| | - Amarine Chancel
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Justin Malcey
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Sébastien Cabrera
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Patrice Fort
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Renato M Maciel
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| |
Collapse
|
3
|
Braine A, Georges F. Emotion in action: When emotions meet motor circuits. Neurosci Biobehav Rev 2023; 155:105475. [PMID: 37996047 DOI: 10.1016/j.neubiorev.2023.105475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
The brain is a remarkably complex organ responsible for a wide range of functions, including the modulation of emotional states and movement. Neuronal circuits are believed to play a crucial role in integrating sensory, cognitive, and emotional information to ultimately guide motor behavior. Over the years, numerous studies employing diverse techniques such as electrophysiology, imaging, and optogenetics have revealed a complex network of neural circuits involved in the regulation of emotional or motor processes. Emotions can exert a substantial influence on motor performance, encompassing both everyday activities and pathological conditions. The aim of this review is to explore how emotional states can shape movements by connecting the neural circuits for emotional processing to motor neural circuits. We first provide a comprehensive overview of the impact of different emotional states on motor control in humans and rodents. In line with behavioral studies, we set out to identify emotion-related structures capable of modulating motor output, behaviorally and anatomically. Neuronal circuits involved in emotional processing are extensively connected to the motor system. These circuits can drive emotional behavior, essential for survival, but can also continuously shape ongoing movement. In summary, the investigation of the intricate relationship between emotion and movement offers valuable insights into human behavior, including opportunities to enhance performance, and holds promise for improving mental and physical health. This review integrates findings from multiple scientific approaches, including anatomical tracing, circuit-based dissection, and behavioral studies, conducted in both animal and human subjects. By incorporating these different methodologies, we aim to present a comprehensive overview of the current understanding of the emotional modulation of movement in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Anaelle Braine
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | | |
Collapse
|
4
|
Vetrivelan R, Bandaru SS. Neural Control of REM Sleep and Motor Atonia: Current Perspectives. Curr Neurol Neurosci Rep 2023; 23:907-923. [PMID: 38060134 DOI: 10.1007/s11910-023-01322-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE OF REVIEW Since the formal discovery of rapid eye movement (REM) sleep in 1953, we have gained a vast amount of knowledge regarding the specific populations of neurons, their connections, and synaptic mechanisms regulating this stage of sleep and its accompanying features. This article discusses REM sleep circuits and their dysfunction, specifically emphasizing recent studies using conditional genetic tools. RECENT FINDINGS Sublaterodorsal nucleus (SLD) in the dorsolateral pons, especially the glutamatergic subpopulation in this region (SLDGlut), are shown to be indispensable for REM sleep. These neurons appear to be single REM generators in the rodent brain and may initiate and orchestrate all REM sleep events, including cortical and hippocampal activation and muscle atonia through distinct pathways. However, several cell groups in the brainstem and hypothalamus may influence SLDGlut neuron activity, thereby modulating REM sleep timing, amounts, and architecture. Damage to SLDGlut neurons or their projections involved in muscle atonia leads to REM behavior disorder, whereas the abnormal activation of this pathway during wakefulness may underlie cataplexy in narcolepsy. Despite some opposing views, it has become evident that SLDGlut neurons are the sole generators of REM sleep and its associated characteristics. Further research should prioritize a deeper understanding of their cellular, synaptic, and molecular properties, as well as the mechanisms that trigger their activation during cataplexy and make them susceptible in RBD.
Collapse
Affiliation(s)
- Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
| | - Sathyajit Sai Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| |
Collapse
|
5
|
Dergacheva O, Fleury-Curado T, Polotsky VY, Kay M, Jain V, Mendelowitz D. GABA and glycine neurons from the ventral medullary region inhibit hypoglossal motoneurons. Sleep 2021; 43:5674942. [PMID: 31832664 DOI: 10.1093/sleep/zsz301] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common disorder characterized by repetitive sleep-related losses of upper airway patency that occur most frequently during rapid eye movement (REM) sleep. Hypoglossal motoneurons play a key role in regulating upper airway muscle tone and patency during sleep. REM sleep activates GABA and glycine neurons in the ventral medulla (VM) to induce cortical desynchronization and skeletal muscle atonia during REM sleep; however, the role of this brain region in modulating hypoglossal motor activity is unknown. We combined optogenetic and chemogenetic approaches with in-vitro and in-vivo electrophysiology, respectfully, in GAD2-Cre mice of both sexes to test the hypothesis that VM GABA/glycine neurons control the activity of hypoglossal motoneurons and tongue muscles. Here, we show that there is a pathway originating from GABA/glycine neurons in the VM that monosynaptically inhibits brainstem hypoglossal motoneurons innervating both tongue protruder genioglossus (GMNs) and retractor (RMNs) muscles. Optogenetic activation of ChR2-expressing fibers induced a greater postsynaptic inhibition in RMNs than in GMNs. In-vivo chemogenetic activation of VM GABA/glycine neurons produced an inhibitory effect on tongue electromyographic (EMG) activity, decreasing both the amplitude and duration of inspiratory-related EMG bursts without any change in respiratory rate. These results indicate that activation of GABA/glycine neurons from the VM inhibits tongue muscles via a direct pathway to both GMNs and RMNs. This inhibition may play a role in REM sleep associated upper airway obstructions that occur in patients with OSA.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, the George Washington University, Washington, DC
| | - Thomaz Fleury-Curado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Matthew Kay
- Department of Biomedical Engineering, the George Washington University, Washington, DC
| | - Vivek Jain
- Department of Medicine, the George Washington University, Washington, DC
| | - David Mendelowitz
- Department of Pharmacology and Physiology, the George Washington University, Washington, DC
| |
Collapse
|
6
|
A Discrete Glycinergic Neuronal Population in the Ventromedial Medulla That Induces Muscle Atonia during REM Sleep and Cataplexy in Mice. J Neurosci 2021; 41:1582-1596. [PMID: 33372061 DOI: 10.1523/jneurosci.0688-20.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
During rapid eye movement (REM) sleep, anti-gravity muscle tone and bodily movements are mostly absent, because somatic motoneurons are inhibited by descending inhibitory pathways. Recent studies showed that glycine/GABA neurons in the ventromedial medulla (VMM; GlyVMM neurons) play an important role in generating muscle atonia during REM sleep (REM-atonia). However, how these REM-atonia-inducing neurons interconnect with other neuronal populations has been unknown. In the present study, we first identified a specific subpopulation of GlyVMM neurons that play an important role in induction of REM-atonia by virus vector-mediated tracing in male mice in which glycinergic neurons expressed Cre recombinase. We found these neurons receive direct synaptic input from neurons in several brain stem regions, including glutamatergic neurons in the sublaterodorsal tegmental nucleus (SLD; GluSLD neurons). Silencing this circuit by specifically expressing tetanus toxin light chain (TeTNLC) resulted in REM sleep without atonia. This manipulation also caused a marked decrease in time spent in cataplexy-like episodes (CLEs) when applied to narcoleptic orexin-ataxin-3 mice. We also showed that GlyVMM neurons play an important role in maintenance of sleep. This present study identified a population of glycinergic neurons in the VMM that are commonly involved in REM-atonia and cataplexy.SIGNIFICANCE STATEMENT We identified a population of glycinergic neurons in the ventral medulla that plays an important role in inducing muscle atonia during rapid eye movement (REM) sleep. It sends axonal projections almost exclusively to motoneurons in the spinal cord and brain stem except to those that innervate extraocular muscles, while other glycinergic neurons in the same region also send projections to other regions including monoaminergic nuclei. Furthermore, these neurons receive direct inputs from several brainstem regions including glutamatergic neurons in the sublaterodorsal tegmental nucleus (SLD). Genetic silencing of this pathway resulted in REM sleep without atonia and a decrease of cataplexy when applied to narcoleptic mice. This work identified a neural population involved in generating muscle atonia during REM sleep and cataplexy.
Collapse
|
7
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
8
|
Park SH, Weber F. Neural and Homeostatic Regulation of REM Sleep. Front Psychol 2020; 11:1662. [PMID: 32793050 PMCID: PMC7385183 DOI: 10.3389/fpsyg.2020.01662] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rapid eye movement (REM) sleep is a distinct, homeostatically controlled brain state characterized by an activated electroencephalogram (EEG) in combination with paralysis of skeletal muscles and is associated with vivid dreaming. Understanding how REM sleep is controlled requires identification of the neural circuits underlying its initiation and maintenance, and delineation of the homeostatic processes regulating its expression on multiple timescales. Soon after its discovery in humans in 1953, the pons was demonstrated to be necessary and sufficient for the generation of REM sleep. But, especially within the last decade, researchers have identified further neural populations in the hypothalamus, midbrain, and medulla that regulate REM sleep by either promoting or suppressing this brain state. The discovery of these populations was greatly facilitated by the availability of novel technologies for the dissection of neural circuits. Recent quantitative models integrate findings about the activity and connectivity of key neurons and knowledge about homeostatic mechanisms to explain the dynamics underlying the recurrence of REM sleep. For the future, combining quantitative with experimental approaches to directly test model predictions and to refine existing models will greatly advance our understanding of the neural and homeostatic processes governing the regulation of REM sleep.
Collapse
Affiliation(s)
| | - Franz Weber
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
9
|
Héricé C, Patel AA, Sakata S. Circuit mechanisms and computational models of REM sleep. Neurosci Res 2018; 140:77-92. [PMID: 30118737 PMCID: PMC6403104 DOI: 10.1016/j.neures.2018.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/03/2018] [Accepted: 07/10/2018] [Indexed: 01/31/2023]
Abstract
REM sleep was discovered in the 1950s. Many hypothalamic and brainstem areas have been found to contribute to REM sleep. An up-to-date picture of REM-sleep-regulating circuits is reviewed. A brief overview of computational models for REM sleep regulation is provided. Outstanding issues for future studies are discussed.
Rapid eye movement (REM) sleep or paradoxical sleep is an elusive behavioral state. Since its discovery in the 1950s, our knowledge of the neuroanatomy, neurotransmitters and neuropeptides underlying REM sleep regulation has continually evolved in parallel with the development of novel technologies. Although the pons was initially discovered to be responsible for REM sleep, it has since been revealed that many components in the hypothalamus, midbrain, pons, and medulla also contribute to REM sleep. In this review, we first provide an up-to-date overview of REM sleep-regulating circuits in the brainstem and hypothalamus by summarizing experimental evidence from neuroanatomical, neurophysiological and gain- and loss-of-function studies. Second, because quantitative approaches are essential for understanding the complexity of REM sleep-regulating circuits and because mathematical models have provided valuable insights into the dynamics underlying REM sleep genesis and maintenance, we summarize computational studies of the sleep-wake cycle, with an emphasis on REM sleep regulation. Finally, we discuss outstanding issues for future studies.
Collapse
Affiliation(s)
- Charlotte Héricé
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Amisha A Patel
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Shuzo Sakata
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
10
|
Ventromedial medulla inhibitory neuron inactivation induces REM sleep without atonia and REM sleep behavior disorder. Nat Commun 2018; 9:504. [PMID: 29402935 PMCID: PMC5799338 DOI: 10.1038/s41467-017-02761-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022] Open
Abstract
Despite decades of research, there is a persistent debate regarding the localization of GABA/glycine neurons responsible for hyperpolarizing somatic motoneurons during paradoxical (or REM) sleep (PS), resulting in the loss of muscle tone during this sleep state. Combining complementary neuroanatomical approaches in rats, we first show that these inhibitory neurons are localized within the ventromedial medulla (vmM) rather than within the spinal cord. We then demonstrate their functional role in PS expression through local injections of adeno-associated virus carrying specific short-hairpin RNA in order to chronically impair inhibitory neurotransmission from vmM. After such selective genetic inactivation, rats display PS without atonia associated with abnormal and violent motor activity, concomitant with a small reduction of daily PS quantity. These symptoms closely mimic human REM sleep behavior disorder (RBD), a prodromal parasomnia of synucleinopathies. Our findings demonstrate the crucial role of GABA/glycine inhibitory vmM neurons in muscle atonia during PS and highlight a candidate brain region that can be susceptible to α-synuclein-dependent degeneration in RBD patients.
Collapse
|
11
|
Bardóczi Z, Pál B, Kőszeghy Á, Wilheim T, Watanabe M, Záborszky L, Liposits Z, Kalló I. Glycinergic Input to the Mouse Basal Forebrain Cholinergic Neurons. J Neurosci 2017; 37:9534-9549. [PMID: 28874448 PMCID: PMC5618268 DOI: 10.1523/jneurosci.3348-16.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 11/21/2022] Open
Abstract
The basal forebrain (BF) receives afferents from brainstem ascending pathways, which has been implicated first by Moruzzi and Magoun (1949) to induce forebrain activation and cortical arousal/waking behavior; however, it is very little known about how brainstem inhibitory inputs affect cholinergic functions. In the current study, glycine, a major inhibitory neurotransmitter of brainstem neurons, and gliotransmitter of local glial cells, was tested for potential interaction with BF cholinergic (BFC) neurons in male mice. In the BF, glycine receptor α subunit-immunoreactive (IR) sites were localized in choline acetyltransferase (ChAT)-IR neurons. The effect of glycine on BFC neurons was demonstrated by bicuculline-resistant, strychnine-sensitive spontaneous IPSCs (sIPSCs; 0.81 ± 0.25 × 10-1 Hz) recorded in whole-cell conditions. Potential neuronal as well as glial sources of glycine were indicated in the extracellular space of cholinergic neurons by glycine transporter type 1 (GLYT1)- and GLYT2-IR processes found in apposition to ChAT-IR cells. Ultrastructural analyses identified synapses of GLYT2-positive axon terminals on ChAT-IR neurons, as well as GLYT1-positive astroglial processes, which were localized in the vicinity of synapses of ChAT-IR neurons. The brainstem raphe magnus was determined to be a major source of glycinergic axons traced retrogradely from the BF. Our results indicate a direct effect of glycine on BFC neurons. Furthermore, the presence of high levels of plasma membrane glycine transporters in the vicinity of cholinergic neurons suggests a tight control of extracellular glycine in the BF.SIGNIFICANCE STATEMENT Basal forebrain cholinergic (BFC) neurons receive various activating inputs from specific brainstem areas and channel this information to the cortex via multiple projections. So far, very little is known about inhibitory brainstem afferents to the BF. The current study established glycine as a major regulator of BFC neurons by (1) identifying glycinergic neurons in the brainstem projecting to the BF, (2) showing glycine receptor α subunit-immunoreactive (IR) sites in choline acetyltransferase (ChAT)-IR neurons, (3) demonstrating glycine transporter type 2 (GLYT2)-positive axon terminals synapsing on ChAT-IR neurons, and (4) localizing GLYT1-positive astroglial processes in the vicinity of synapses of ChAT-IR neurons. The effect of glycine on BFC neurons was demonstrated by bicuculline-resistant, strychnine-sensitive spontaneous IPSCs recorded in whole-cell conditions.
Collapse
Affiliation(s)
- Zsuzsanna Bardóczi
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
- Semmelweis University, School of PH.D. Studies, 1085, Budapest, Hungary
| | - Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
| | - Áron Kőszeghy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
| | - Tamás Wilheim
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, 1083, Budapest, Hungary
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo 060-8638, Japan
| | - László Záborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, Newark, New Jersey 07102, and
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, 1083, Budapest, Hungary
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, HAS, 1083, Budapest, Hungary,
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, 1083, Budapest, Hungary
| |
Collapse
|
12
|
Luppi PH, Peyron C, Fort P. Not a single but multiple populations of GABAergic neurons control sleep. Sleep Med Rev 2017; 32:85-94. [DOI: 10.1016/j.smrv.2016.03.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 12/15/2022]
|
13
|
Chen MC, Vetrivelan R, Guo CN, Chang C, Fuller PM, Lu J. Ventral medullary control of rapid eye movement sleep and atonia. Exp Neurol 2017; 290:53-62. [PMID: 28077261 DOI: 10.1016/j.expneurol.2017.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/24/2016] [Accepted: 01/04/2017] [Indexed: 11/27/2022]
Abstract
Discrete populations of neurons at multiple levels of the brainstem control rapid eye movement (REM) sleep and the accompanying loss of postural muscle tone, or atonia. The specific contributions of these brainstem cell populations to REM sleep control remains incompletely understood. Here we show in rats that viral vector-based lesions of the ventromedial medulla at a level rostral to the inferior olive (pSOM) produced violent myoclonic twitches and abnormal electromyographic spikes, but not complete loss of tonic atonia, during REM sleep. Motor tone during non-REM (NREM) sleep was unaffected in these same animals. Acute chemogenetic activation of pSOM neurons in rats robustly and selectively suppressed REM sleep but not NREM sleep. Similar lesions targeting the more rostral ventromedial medulla (RVM) did not affect sleep or atonia, while chemogenetic stimulation of the RVM produced wakefulness and reduced sleep. Finally, selective activation of vesicular GABA transporter (VGAT) pSOM neurons in mice produced complete suppression of REM sleep whereas their loss increased EMG spikes during REM sleep. These results reveal a key contribution of the pSOM and specifically the VGAT+ neurons in this region in REM sleep and motor control.
Collapse
Affiliation(s)
- Michael C Chen
- Beth Israel Deaconess Medical Center and Harvard Medical School, Department of Neurology, Division of Sleep Medicine, Boston, MA 02115, USA
| | - Ramalingam Vetrivelan
- Beth Israel Deaconess Medical Center and Harvard Medical School, Department of Neurology, Division of Sleep Medicine, Boston, MA 02115, USA
| | - Chun-Ni Guo
- Department of Neurology, Shanghai First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Catie Chang
- Advanced Magnetic Resonance Imaging Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patrick M Fuller
- Beth Israel Deaconess Medical Center and Harvard Medical School, Department of Neurology, Division of Sleep Medicine, Boston, MA 02115, USA
| | - Jun Lu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Department of Neurology, Division of Sleep Medicine, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Arrigoni E, Chen MC, Fuller PM. The anatomical, cellular and synaptic basis of motor atonia during rapid eye movement sleep. J Physiol 2016; 594:5391-414. [PMID: 27060683 DOI: 10.1113/jp271324] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/02/2016] [Indexed: 01/14/2023] Open
Abstract
Rapid eye movement (REM) sleep is a recurring part of the sleep-wake cycle characterized by fast, desynchronized rhythms in the electroencephalogram (EEG), hippocampal theta activity, rapid eye movements, autonomic activation and loss of postural muscle tone (atonia). The brain circuitry governing REM sleep is located in the pontine and medullary brainstem and includes ascending and descending projections that regulate the EEG and motor components of REM sleep. The descending signal for postural muscle atonia during REM sleep is thought to originate from glutamatergic neurons of the sublaterodorsal nucleus (SLD), which in turn activate glycinergic pre-motor neurons in the spinal cord and/or ventromedial medulla to inhibit motor neurons. Despite work over the past two decades on many neurotransmitter systems that regulate the SLD, gaps remain in our knowledge of the synaptic basis by which SLD REM neurons are regulated and in turn produce REM sleep atonia. Elucidating the anatomical, cellular and synaptic basis of REM sleep atonia control is a critical step for treating many sleep-related disorders including obstructive sleep apnoea (apnea), REM sleep behaviour disorder (RBD) and narcolepsy with cataplexy.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| | - Michael C Chen
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
15
|
Leiras R, Martín-Cora F, Velo P, Liste T, Canedo A. Cat's medullary reticulospinal and subnucleus reticularis dorsalis noxious neurons form a coupled neural circuit through collaterals of descending axons. J Neurophysiol 2015; 115:324-44. [PMID: 26581870 DOI: 10.1152/jn.00603.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/04/2015] [Indexed: 12/17/2022] Open
Abstract
Animals and human beings sense and react to real/potential dangerous stimuli. However, the supraspinal mechanisms relating noxious sensing and nocifensive behavior are mostly unknown. The collateralization and spatial organization of interrelated neurons are important determinants of coordinated network function. Here we electrophysiologically studied medial medullary reticulospinal neurons (mMRF-RSNs) antidromically identified from the cervical cord of anesthetized cats and found that 1) more than 40% (79/183) of the sampled mMRF-RSNs emitted bifurcating axons running within the dorsolateral (DLF) and ventromedial (VMF) ipsilateral fascicles; 2) more than 50% (78/151) of the tested mMRF-RSNs with axons running in the VMF collateralized to the subnucleus reticularis dorsalis (SRD) that also sent ipsilateral descending fibers bifurcating within the DLF and the VMF. This percentage of mMRF collateralization to the SRD increased to more than 81% (53/65) when considering the subpopulation of mMRF-RSNs responsive to noxiously heating the skin; 3) reciprocal monosynaptic excitatory relationships were electrophysiologically demonstrated between noxious sensitive mMRF-RSNs and SRD cells; and 4) injection of the anterograde tracer Phaseolus vulgaris leucoagglutinin evidenced mMRF to SRD and SRD to mMRF projections contacting the soma and proximal dendrites. The data demonstrated a SRD-mMRF network interconnected mainly through collaterals of descending axons running within the VMF, with the subset of noxious sensitive cells forming a reverberating circuit probably amplifying mutual outputs simultaneously regulating motor activity and spinal noxious afferent input. The results provide evidence that noxious stimulation positively engages a reticular SRD-mMRF-SRD network involved in pain-sensory-to-motor transformation and modulation.
Collapse
Affiliation(s)
- Roberto Leiras
- Department of Physiology, Faculty Medicine, University Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisco Martín-Cora
- Department of Physiology, Faculty Medicine, University Santiago de Compostela, Santiago de Compostela, Spain
| | - Patricia Velo
- Department of Physiology, Faculty Medicine, University Santiago de Compostela, Santiago de Compostela, Spain
| | - Tania Liste
- Department of Physiology, Faculty Medicine, University Santiago de Compostela, Santiago de Compostela, Spain
| | - Antonio Canedo
- Department of Physiology, Faculty Medicine, University Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
16
|
The sleep-promoting and hypothermic effects of glycine are mediated by NMDA receptors in the suprachiasmatic nucleus. Neuropsychopharmacology 2015; 40:1405-16. [PMID: 25533534 PMCID: PMC4397399 DOI: 10.1038/npp.2014.326] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 11/17/2014] [Accepted: 12/01/2014] [Indexed: 01/08/2023]
Abstract
The use of glycine as a therapeutic option for improving sleep quality is a novel and safe approach. However, despite clinical evidence of its efficacy, the details of its mechanism remain poorly understood. In this study, we investigated the site of action and sleep-promoting mechanisms of glycine in rats. In acute sleep disturbance, oral administration of glycine-induced non-rapid eye movement (REM) sleep and shortened NREM sleep latency with a simultaneous decrease in core temperature. Oral and intracerebroventricular injection of glycine elevated cutaneous blood flow (CBF) at the plantar surface in a dose-dependent manner, resulting in heat loss. Pretreatment with N-methyl-D-aspartate (NMDA) receptor antagonists AP5 and CGP78608 but not the glycine receptor antagonist strychnine inhibited the CBF increase caused by glycine injection into the brain. Induction of c-Fos expression was observed in the hypothalamic nuclei, including the medial preoptic area (MPO) and the suprachiasmatic nucleus (SCN) shell after glycine administration. Bilateral microinjection of glycine into the SCN elevated CBF in a dose-dependent manner, whereas no effect was observed when glycine was injected into the MPO and dorsal subparaventricular zone. In addition, microinjection of D-serine into the SCN also increased CBF, whereas these effects were blocked in the presence of L-701324. SCN ablation completely abolished the sleep-promoting and hypothermic effects of glycine. These data suggest that exogenous glycine promotes sleep via peripheral vasodilatation through the activation of NMDA receptors in the SCN shell.
Collapse
|
17
|
Serotonergic transmission after spinal cord injury. J Neural Transm (Vienna) 2014; 122:279-95. [PMID: 24866695 DOI: 10.1007/s00702-014-1241-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/06/2014] [Indexed: 12/27/2022]
Abstract
Changes in descending serotonergic innervation of spinal neural activity have been implicated in symptoms of paralysis, spasticity, sensory disturbances and pain following spinal cord injury (SCI). Serotonergic neurons possess an enhanced ability to regenerate or sprout after many types of injury, including SCI. Current research suggests that serotonine (5-HT) release within the ventral horn of the spinal cord plays a critical role in motor function, and activation of 5-HT receptors mediates locomotor control. 5-HT originating from the brain stem inhibits sensory afferent transmission and associated spinal reflexes; by abolishing 5-HT innervation SCI leads to a disinhibition of sensory transmission. 5-HT denervation supersensitivity is one of the key mechanisms underlying the increased motoneuron excitability that occurs after SCI, and this hyperexcitability has been demonstrated to underlie the pathogenesis of spasticity after SCI. Moreover, emerging evidence implicates serotonergic descending facilitatory pathways from the brainstem to the spinal cord in the maintenance of pathologic pain. There are functional relevant connections between the descending serotonergic system from the rostral ventromedial medulla in the brainstem, the 5-HT receptors in the spinal dorsal horn, and the descending pain facilitation after tissue and nerve injury. This narrative review focussed on the most important studies that have investigated the above-mentioned effects of impaired 5-HT-transmission in humans after SCI. We also briefly discussed the promising therapeutical approaches with serotonergic drugs, monoclonal antibodies and intraspinal cell transplantation.
Collapse
|
18
|
Holstege G. The periaqueductal gray controls brainstem emotional motor systems including respiration. PROGRESS IN BRAIN RESEARCH 2014; 209:379-405. [PMID: 24746059 DOI: 10.1016/b978-0-444-63274-6.00020-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Respiration is a motor system essential for the survival of the individual and of the species. Because of its vital significance, studies on respiration often assume that breathing takes place independent of other motor systems. However, motor systems generating vocalization, coughing, sneezing, vomiting, as well as parturition, ejaculation, and defecation encompass abdominal pressure control, which involves changes in the respiratory pattern. The mesencephalic periaqueductal gray (PAG) controls all these motor systems. It determines the level setting of the whole body by means of its very strong projections to the ventromedial medullary tegmentum, but it also controls the cell groups that generate vocalization, coughing, sneezing, vomiting, as well as respiration. For this control, the PAG maintains very strong connections with the nucleus retroambiguus, which enables it to control abdominal and intrathoracic pressure. In this same context, the PAG also runs the pelvic organs, bladder, uterus, prostate, seminal vesicles, and the distal colon and rectum via its projections to the pelvic organ stimulating center and the pelvic floor stimulating center. These cell groups, via long descending projections, have direct control of the parasympathetic motoneurons in the sacral cord as well as of the somatic motoneurons in the nucleus of Onuf, innervating the pelvic floor. Respiration, therefore, is not a motor system that functions by itself, but is strongly regulated by the same systems that also control the other motor output systems.
Collapse
Affiliation(s)
- Gert Holstege
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia.
| |
Collapse
|
19
|
Chang T, Alexopoulos H, Pettingill P, McMenamin M, Deacon R, Erdelyi F, Szabó G, Buckley CJ, Vincent A. Immunization against GAD induces antibody binding to GAD-independent antigens and brainstem GABAergic neuronal loss. PLoS One 2013; 8:e72921. [PMID: 24058450 PMCID: PMC3776810 DOI: 10.1371/journal.pone.0072921] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/15/2013] [Indexed: 11/18/2022] Open
Abstract
Stiff person syndrome (SPS) is a highly-disabling neurological disorder of the CNS characterized by progressive muscular rigidity and spasms. In approximately 60–80% of patients there are autoantibodies to glutamic acid decarboxylase (GAD), the enzyme that synthesizes gamma-amino butyric acid (GABA), the predominant inhibitory neurotransmitter of the CNS. Although GAD is intracellular, it is thought that autoimmunity to GAD65 may play a role in the development of SPS. To test this hypothesis, we immunized mice, that expressed enhanced green fluorescent protein (EGFP) under the GAD65 promoter, with either GAD65 (n = 13) or phosphate buffered saline (PBS) (n = 13). Immunization with GAD65 resulted in autoantibodies that immunoprecipitated GAD, bound to CNS tissue in a highly characteristic pattern, and surprisingly bound not only to GAD intracellularly but also to the surface of cerebellar neurons in culture. Moreover, immunization resulted in immunoglobulin diffusion into the brainstem, and a partial loss of GAD-EGFP expressing cells in the brainstem. Although immunization with GAD65 did not produce any behavioral abnormality in the mice, the induction of neuronal-surface antibodies and the trend towards loss of GABAergic neurons in the brainstem, supports a role for humoral autoimmunity in the pathogenesis of SPS and suggests that the mechanisms may involve spread to antigens expressed on the surface of these neurons.
Collapse
Affiliation(s)
- Thashi Chang
- Neuroimmunology Group, Weatherall Institute of Molecular Medicine and Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Department of Clinical Medicine, University of Colombo, Colombo, Sri Lanka
| | - Harry Alexopoulos
- Neuroimmunology Group, Weatherall Institute of Molecular Medicine and Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Philippa Pettingill
- Neuroimmunology Group, Weatherall Institute of Molecular Medicine and Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mary McMenamin
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Robert Deacon
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Ferenc Erdelyi
- Department of Gene Technology and Developmental Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gabor Szabó
- Department of Gene Technology and Developmental Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Camilla J. Buckley
- Neuroimmunology Group, Weatherall Institute of Molecular Medicine and Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Angela Vincent
- Neuroimmunology Group, Weatherall Institute of Molecular Medicine and Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Luppi PH, Clément O, Valencia Garcia S, Brischoux F, Fort P. New aspects in the pathophysiology of rapid eye movement sleep behavior disorder: the potential role of glutamate, gamma-aminobutyric acid, and glycine. Sleep Med 2013; 14:714-8. [DOI: 10.1016/j.sleep.2013.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 02/12/2013] [Accepted: 02/14/2013] [Indexed: 10/26/2022]
|
21
|
Luppi PH, Clément O, Fort P. Brainstem structures involved in rapid eye movement sleep behavior disorder. Sleep Biol Rhythms 2013. [DOI: 10.1111/j.1479-8425.2012.00544.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Krenzer M, Lu J, Mayer G, Oertel W. From bench to bed: putative animal models of REM sleep behavior disorder (RBD). J Neural Transm (Vienna) 2013; 120:683-8. [PMID: 23338670 DOI: 10.1007/s00702-012-0965-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 12/19/2012] [Indexed: 11/25/2022]
Abstract
REM behavior disorder (RBD) is a parasomnia characterized by REM sleep without atonia, leading to abnormal and potentially injurious behavior during REM sleep. It is considered one of the most specific predictors of neurodegenerative disorders, such as Parkinson's disease. In this paper, we provide an overview of animal models contributing to our current understanding of REM-associated atonia, and, as a consequence, the pathophysiology of RBD. The generator of REM-associated atonia is located in glutamatergic neurons of the pontine sublaterodorsal nucleus (SLD), as shown in cats, rats and mice. These findings are supported by clinical cases of patients with lesions of the homologous structure in humans. Glutamatergic SLD neurons, presumably in conjunction with others, project to (a) the ventromedial medulla, where they either directly target inhibitory interneurons to alpha motor neurons or are relayed, and (b) the spinal cord directly. At the spinal level, alpha motor neurons are inhibited by GABAergic and glycinergic interneurons. Our current understanding is that lesions of the glutamatergic SLD are the key factor for REM sleep behavior disorder. However, open questions remain, e.g. other features of RBD (such as the typically aggressive dream content) or the frequent progression from idiopathic RBD to neurodegenerative disorders, to name only a few. In order to elucidate these questions, a constant interaction between basic and clinical researchers is required, which might, ultimately, create an early therapeutic window for neurodegenerative disorders.
Collapse
Affiliation(s)
- Martina Krenzer
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany.
| | | | | | | |
Collapse
|
23
|
Barreiro-Iglesias A, Mysiak KS, Adrio F, Rodicio MC, Becker CG, Becker T, Anadón R. Distribution of glycinergic neurons in the brain of glycine transporter-2 transgenic Tg(glyt2:Gfp) adult zebrafish: Relationship to brain-spinal descending systems. J Comp Neurol 2012; 521:389-425. [DOI: 10.1002/cne.23179] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 01/25/2012] [Accepted: 06/21/2012] [Indexed: 12/19/2022]
|
24
|
Du Beau A, Shakya Shrestha S, Bannatyne BA, Jalicy SM, Linnen S, Maxwell DJ. Neurotransmitter phenotypes of descending systems in the rat lumbar spinal cord. Neuroscience 2012; 227:67-79. [PMID: 23018001 DOI: 10.1016/j.neuroscience.2012.09.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/03/2012] [Accepted: 09/14/2012] [Indexed: 12/01/2022]
Abstract
Descending systems from the brain exert a major influence over sensory and motor processes within the spinal cord. Although it is known that many descending systems have an excitatory effect on spinal neurons, there are still gaps in our knowledge regarding the transmitter phenotypes used by them. In this study we investigated transmitter phenotypes of axons in the corticospinal tract (CST); the rubrospinal tract (RST); the lateral component of the vestibulospinal tract (VST); and the reticulospinal tract (ReST). They were labelled anterogradely by stereotaxic injection of the b subunit of cholera toxin (CTb) into the motor cortex, red nucleus, lateral vestibular nucleus and medial longitudinal fascicle (MLF) to label CST, RST, VST and ReST axons respectively. Neurotransmitter content of labelled axons was investigated in lumbar segments by using immunoflurescence; antibodies against vesicular glutamate transporters (VGLUT1 and VGLUT2) were used to identify glutamatergic terminals and the vesicular GABA transporter (VGAT) was used to identify GABA- and glycinergic terminals. The results show that almost all CST (96%) axons contain VGLUT1 whereas almost all RST (97%) and VST (97%) axons contain VGLUT2. Although the majority of ReST axons contain VGLUT2 (59%), a sizable minority contains VGAT (20%) and most of these terminals can be subdivided into those that are GABAergic or those that are glycinergic because only limited evidence for co-localisation was found for the two transmitters. In addition, there is a population of ReST terminals that apparently does not contain markers for the transmitters tested and is not serotoninergic. We can conclude that the CST, RST and VST are 'pure' excitatory systems whereas the ReST consists of a heterogeneous population of excitatory and inhibitory axons. It is anticipated that this information will enable inputs to spinal networks to be defined with greater confidence.
Collapse
Affiliation(s)
- A Du Beau
- Spinal Cord Group, Institute of Neuroscience and Psychology, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | | | | | | | | | | |
Collapse
|
25
|
Inhibitory inputs to four types of spinocerebellar tract neurons in the cat spinal cord. Neuroscience 2012; 226:253-69. [PMID: 22989920 DOI: 10.1016/j.neuroscience.2012.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 09/05/2012] [Accepted: 09/06/2012] [Indexed: 11/24/2022]
Abstract
Spinocerebellar tract neurons are inhibited by various sources of input via pathways activated by descending tracts as well as peripheral afferents. Inhibition may be used to modulate transmission of excitatory information forwarded to the cerebellum. However it may also provide information on the degree of inhibition of motoneurons and on the operation of inhibitory premotor neurons. Our aim was to extend previous comparisons of morphological substrates of excitation of spinocerebellar neurons to inhibitory input. Contacts formed by inhibitory axon terminals were characterised as either GABAergic, glycinergic or both GABAergic/glycinergic by using antibodies against vesicular GABA transporter, glutamic acid decarboxylase and gephyrin. Quantitative analysis revealed the presence of much higher proportions of inhibitory contacts when compared with excitatory contacts on spinal border (SB) neurons. However similar proportions of inhibitory and excitatory contacts were associated with ventral spinocerebellar tract (VSCT) and dorsal spinocerebellar tract neurons located in Clarke's column (ccDSCT) and the dorsal horn (dhDSCT). In all of the cells, the majority of inhibitory terminals were glycinergic. The density of contacts was higher on somata and proximal versus distal dendrites of SB and VSCT neurons but more evenly distributed in ccDSCT and dhDSCT neurons. Variations in the density and distribution of inhibitory contacts found in this study may reflect differences in information on inhibitory processes forwarded by subtypes of spinocerebellar tract neurons to the cerebellum.
Collapse
|
26
|
Abstract
This review summarizes the brain mechanisms controlling sleep and wakefulness. Wakefulness promoting systems cause low-voltage, fast activity in the electroencephalogram (EEG). Multiple interacting neurotransmitter systems in the brain stem, hypothalamus, and basal forebrain converge onto common effector systems in the thalamus and cortex. Sleep results from the inhibition of wake-promoting systems by homeostatic sleep factors such as adenosine and nitric oxide and GABAergic neurons in the preoptic area of the hypothalamus, resulting in large-amplitude, slow EEG oscillations. Local, activity-dependent factors modulate the amplitude and frequency of cortical slow oscillations. Non-rapid-eye-movement (NREM) sleep results in conservation of brain energy and facilitates memory consolidation through the modulation of synaptic weights. Rapid-eye-movement (REM) sleep results from the interaction of brain stem cholinergic, aminergic, and GABAergic neurons which control the activity of glutamatergic reticular formation neurons leading to REM sleep phenomena such as muscle atonia, REMs, dreaming, and cortical activation. Strong activation of limbic regions during REM sleep suggests a role in regulation of emotion. Genetic studies suggest that brain mechanisms controlling waking and NREM sleep are strongly conserved throughout evolution, underscoring their enormous importance for brain function. Sleep disruption interferes with the normal restorative functions of NREM and REM sleep, resulting in disruptions of breathing and cardiovascular function, changes in emotional reactivity, and cognitive impairments in attention, memory, and decision making.
Collapse
Affiliation(s)
- Ritchie E Brown
- Laboratory of Neuroscience, VA Boston Healthcare System and Harvard Medical School, Brockton, Massachusetts 02301, USA
| | | | | | | | | |
Collapse
|
27
|
Hossaini M, Goos JAC, Kohli SK, Holstege JC. Distribution of glycine/GABA neurons in the ventromedial medulla with descending spinal projections and evidence for an ascending glycine/GABA projection. PLoS One 2012; 7:e35293. [PMID: 22558137 PMCID: PMC3340372 DOI: 10.1371/journal.pone.0035293] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/14/2012] [Indexed: 11/23/2022] Open
Abstract
The ventromedial medulla (VM), subdivided in a rostral (RVM) and a caudal (CVM) part, has a powerful influence on the spinal cord. In this study, we have identified the distribution of glycine and GABA containing neurons in the VM with projections to the cervical spinal cord, the lumbar dorsal horn, and the lumbar ventral horn. For this purpose, we have combined retrograde tracing using fluorescent microspheres with fluorescent in situ hybridization (FISH) for glycine transporter 2 (GlyT2) and GAD67 mRNAs to identify glycinergic and/or GABAergic (Gly/GABA) neurons. Since the results obtained with FISH for GlyT2, GAD67, or GlyT2 + GAD67 mRNAs were not significantly different, we concluded that glycine and GABA coexisted in the various projection neurons. After injections in the cervical cord, we found that 29% ± 1 (SEM) of the retrogradely labeled neurons in the VM were Gly/GABA (RVM: 43%; CVM: 21%). After lumbar dorsal horn injections 31% ± 3 of the VM neurons were Gly/GABA (RVM: 45%; CVM: 12%), and after lumbar ventral horn injections 25% ± 2 were Gly/GABA (RVM: 35%; CVM: 17%). In addition, we have identified a novel ascending Gly/GABA pathway originating from neurons in the area around the central canal (CC) throughout the spinal cord and projecting to the RVM, emphasizing the interaction between the ventromedial medulla and the spinal cord. The present study has now firmly established that GABA and glycine are present in many VM neurons that project to the spinal cord. These neurons strongly influence spinal processing, most notably the inhibition of nociceptive transmission.
Collapse
Affiliation(s)
- Mehdi Hossaini
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jacqueline A. C. Goos
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Somesh K. Kohli
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan C. Holstege
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
A MusD retrotransposon insertion in the mouse Slc6a5 gene causes alterations in neuromuscular junction maturation and behavioral phenotypes. PLoS One 2012; 7:e30217. [PMID: 22272310 PMCID: PMC3260239 DOI: 10.1371/journal.pone.0030217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 12/15/2011] [Indexed: 11/20/2022] Open
Abstract
Glycine is the major inhibitory neurotransmitter in the spinal cord and some brain regions. The presynaptic glycine transporter, GlyT2, is required for sustained glycinergic transmission through presynaptic reuptake and recycling of glycine. Mutations in SLC6A5, encoding GlyT2, cause hereditary hyperekplexia in humans, and similar phenotypes in knock-out mice, and variants are associated with schizophrenia. We identified a spontaneous mutation in mouse Slc6a5, caused by a MusD retrotransposon insertion. The GlyT2 protein is undetectable in homozygous mutants, indicating a null allele. Homozygous mutant mice are normal at birth, but develop handling-induced spasms at five days of age, and only survive for two weeks, but allow the study of early activity-regulated developmental processes. At the neuromuscular junction, synapse elimination and the switch from embryonic to adult acetylcholine receptor subunits are hastened, consistent with a presumed increase in motor neuron activity, and transcription of acetylcholine receptors is elevated. Heterozygous mice, which show no reduction in lifespan but nonetheless have reduced levels of GlyT2, have a normal thermal sensitivity with the hot-plate test, but differences in repetitive grooming and decreased sleep time with home-cage monitoring. Open-field and elevated plus-maze tests did not detect anxiety-like behaviors; however, the latter showed a hyperactivity phenotype. Importantly, grooming and hyperactivity are observed in mouse schizophrenia models. Thus, mutations in Slc6a5 show changes in neuromuscular junction development as homozygotes, and behavioral phenotypes as heterozygotes, indicating their usefulness for studies related to glycinergic dysfunction.
Collapse
|
29
|
Valle-Maroto S, Fernández-López B, Villar-Cerviño V, Barreiro-Iglesias A, Anadón R, Rodicio MC. Inhibitory descending rhombencephalic projections in larval sea lamprey. Neuroscience 2011; 194:1-10. [DOI: 10.1016/j.neuroscience.2011.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/29/2011] [Accepted: 08/06/2011] [Indexed: 10/17/2022]
|
30
|
Murray KC, Stephens MJ, Rank M, D'Amico J, Gorassini MA, Bennett DJ. Polysynaptic excitatory postsynaptic potentials that trigger spasms after spinal cord injury in rats are inhibited by 5-HT1B and 5-HT1F receptors. J Neurophysiol 2011; 106:925-43. [PMID: 21653728 PMCID: PMC3154834 DOI: 10.1152/jn.01011.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 05/26/2011] [Indexed: 02/07/2023] Open
Abstract
Sensory afferent transmission and associated spinal reflexes are normally inhibited by serotonin (5-HT) derived from the brain stem. Spinal cord injury (SCI) that eliminates this 5-HT innervation leads to a disinhibition of sensory transmission and a consequent emergence of unusually long polysynaptic excitatory postsynaptic potentials (EPSPs) in motoneurons. These EPSPs play a critical role in triggering long polysynaptic reflexes (LPRs) that initiate muscles spasms. In the present study we examined which 5-HT receptors modulate the EPSPs and whether these receptors adapt to a loss of 5-HT after chronic spinal transection in rats. The EPSPs and associated LPRs recorded in vitro in spinal cords from chronic spinal rats were consistently inhibited by 5-HT(1B) or 5-HT(1F) receptor agonists, including zolmitriptan (5-HT(1B/1D/1F)) and LY344864 (5-HT(1F)), with a sigmoidal dose-response relation, from which we computed the 50% inhibition (EC(50)) and potency (-log EC(50)). The potencies of 5-HT receptor agonists were highly correlated with their binding affinity to 5-HT(1B) and 5-HT(1F) receptors, and not to other 5-HT receptors. Zolmitriptan also inhibited the LPRs and general muscle spasms recorded in vivo in the awake chronic spinal rat. The 5-HT(1B) receptor antagonists SB216641 and GR127935 and the inverse agonist SB224289 reduced the inhibition of LPRs by 5-HT(1B) agonists (zolmitriptan). However, when applied alone, SB224289, SB216641, and GR127935 had no effect on the LPRs, indicating that 5-HT(1B) receptors do not adapt to chronic injury, remaining silent, without constitutive activity. The reduction in EPSPs with zolmitriptan unmasked a large glycine-mediated inhibitory postsynaptic current (IPSC) after SCI. This IPSC and associated chloride current reversed at -73 mV, slightly below the resting membrane potential. Zolmitriptan did not change motoneuron properties. Our results demonstrate that 5-HT(1B/1F) agonists, such as zolmitriptan, can restore inhibition of sensory transmission after SCI without affecting general motoneuron function and thus may serve as a novel class of antispastic drugs.
Collapse
Affiliation(s)
- Katherine C Murray
- Centre for Neuroscience, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
31
|
Luppi PH, Clément O, Sapin E, Gervasoni D, Peyron C, Léger L, Salvert D, Fort P. The neuronal network responsible for paradoxical sleep and its dysfunctions causing narcolepsy and rapid eye movement (REM) behavior disorder. Sleep Med Rev 2011; 15:153-63. [PMID: 21115377 DOI: 10.1016/j.smrv.2010.08.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 08/11/2010] [Accepted: 08/11/2010] [Indexed: 02/02/2023]
Affiliation(s)
- Pierre-Hervé Luppi
- UMR5167 CNRS, Institut Fédératif des Neurosciences de Lyon (IFR 19), Univ Lyon 1, Université de Lyon, Lyon, France.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- Adrian R Morrison
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
|
34
|
|
35
|
Murray KC, Stephens MJ, Ballou EW, Heckman CJ, Bennett DJ. Motoneuron excitability and muscle spasms are regulated by 5-HT2B and 5-HT2C receptor activity. J Neurophysiol 2010; 105:731-48. [PMID: 20980537 DOI: 10.1152/jn.00774.2010] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immediately after spinal cord injury (SCI), a devastating paralysis results from the loss of brain stem and cortical innervation of spinal neurons that control movement, including a loss of serotonergic (5-HT) innervation of motoneurons. Over time, motoneurons recover from denervation and function autonomously, exhibiting large persistent calcium currents (Ca PICs) that both help with functional recovery and contribute to uncontrolled muscle spasms. Here we systematically evaluated which 5-HT receptor subtypes influence PICs and spasms after injury. Spasms were quantified by recording the long-lasting reflexes (LLRs) on ventral roots in response to dorsal root stimulation, in the chronic spinal rat, in vitro. Ca PICs were quantified by intracellular recording in synaptically isolated motoneurons. Application of agonists selective to 5-HT(2B) and 5-HT(2C) receptors (including BW723C86) significantly increased the LLRs and associated Ca PICs, whereas application of agonists to 5-HT(1), 5-HT(2A), 5-HT(3), or 5-HT(4/5/6/7) receptors (e.g., 8-OH-DPAT) did not. The 5-HT(2) receptor agonist-induced increases in LLRs were dose dependent, with doses for 50% effects (EC(50)) highly correlated with published doses for agonist receptor binding (K(i)) at 5-HT(2B) and 5-HT(2C) receptors. Application of selective antagonists to 5-HT(2B) (e.g., RS127445) and 5-HT(2C) (SB242084) receptors inhibited the agonist-induced increase in LLR. However, antagonists that are known to specifically be neutral antagonists at 5-HT(2B/C) receptors (e.g., RS127445) had no effect when given by themselves, indicating that these receptors were not activated by residual 5-HT in the spinal cord. In contrast, inverse agonists (such as SB206553) that block constitutive activity at 5-HT(2B) or 5-HT(2C) receptors markedly reduced the LLRs, indicating the presence of constitutive activity in these receptors. 5-HT(2B) or 5-HT(2C) receptors were confirmed to be on motoneurons by immunolabeling. In summary, 5-HT(2B) and 5-HT(2C) receptors on motoneurons become constitutively active after injury and ultimately contribute to recovery of motoneuron function and emergence of spasms.
Collapse
Affiliation(s)
- Katherine C Murray
- Centre for Neuroscience, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
36
|
Lai YY, Kodama T, Schenkel E, Siegel JM. Behavioral response and transmitter release during atonia elicited by medial medullary stimulation. J Neurophysiol 2010; 104:2024-33. [PMID: 20668280 DOI: 10.1152/jn.00528.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of the medial medulla is responsible for rapid eye movement (REM) sleep atonia and cataplexy. Dysfunction can cause REM sleep behavior disorder and other motor pathologies. Here we report the behavioral effects of stimulation of the nucleus gigantocellularis (NGC) and nucleus magnocellularis (NMC) in unrestrained cats. In waking, 62% of the medial medullary stimulation sites suppressed muscle tone. In contrast, stimulation at all sites, including sites where stimulation produced no change or increased muscle tone in waking, produced decreased muscle tone during slow-wave sleep. In the decerebrate cat electrical stimulation of the NGC increased glycine and decreased norepinephrine (NE) release in the lumbar ventral horn, with no change in γ-aminobutyric acid (GABA) or serotonin (5-HT) release. Stimulation of the NMC increased both glycine and GABA release and also decreased both NE and 5-HT release in the ventral horn. Glutamate levels in the ventral horn were not changed by either NGC or NMC stimulation. We conclude that NGC and NMC play neurochemically distinct but synergistic roles in the modulation of motor activity across the sleep-wake cycle via a combination of increased release of glycine and GABA and decreased release of 5-HT and NE. Stimulation of the medial medulla that elicited muscle tone suppression also triggered rapid eye movements, but never produced the phasic twitches that characterize REM sleep, indicating that the twitching and rapid eye movement generators of REM sleep have separate brain stem substrates.
Collapse
Affiliation(s)
- Yuan-Yang Lai
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System Sepulveda, North Hills, CA 91343, USA.
| | | | | | | |
Collapse
|
37
|
Co-localisation of markers for glycinergic and GABAergic neurones in rat nucleus of the solitary tract: implications for co-transmission. J Chem Neuroanat 2010; 40:160-76. [PMID: 20434539 DOI: 10.1016/j.jchemneu.2010.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 04/21/2010] [Accepted: 04/22/2010] [Indexed: 01/12/2023]
Abstract
Immunoreactive structures visualised with antibodies to glycine were prominent in areas of the nucleus of the solitary tract (NTS) surrounding the tractus solitarius, but scarcer in medial and ventral areas of the nucleus. This contrasted with a higher density, more homogenous distribution of structures labelled for gamma-aminobutyric acid (GABA). Immunolabelling of adjacent semi-thin sections nonetheless indicated a close correspondence between cells and puncta labelled by glycine and GABA antisera in certain NTS areas. With post-embedding electron microscopic immunolabelling, synaptic terminals with high, presumed transmitter levels of glycine were discriminated from terminals containing low, metabolic levels by quantitative analysis of gold particle labelling densities. In a random sample of terminals, 28.5% qualified on this basis as glycinergic (compared to 44.4% GABAergic); these glycinergic terminals targeted mainly dendritic structures and contained pleomorphic vesicles and symmetrical synapses. Serial section analysis revealed few terminals (5.2%) immunoreactive for glycine alone, with 82% of glycinergic terminals also containing high levels of GABA immunoreactivity. No evidence for co-localisation of glycine and glutamate was found. Light, confocal and electron microscopic labelling with antibodies to proteins specific for glycine and GABA synthesis, release and uptake confirmed that glycinergic terminals also containing GABA are found predominantly in more lateral areas of NTS, despite glycine receptors and the 'glial' glycine transporter (GLYT1) being expressed throughout all areas of the nucleus. The data suggest that synaptic terminals in certain functionally distinct areas of NTS co-release both inhibitory amino acids, which may account for the previously reported differential inhibitory effects of glycine and GABA on NTS neurones.
Collapse
|
38
|
Vetrivelan R, Fuller PM, Tong Q, Lu J. Medullary circuitry regulating rapid eye movement sleep and motor atonia. J Neurosci 2009; 29:9361-9. [PMID: 19625526 PMCID: PMC2758912 DOI: 10.1523/jneurosci.0737-09.2009] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 06/22/2009] [Accepted: 06/23/2009] [Indexed: 11/21/2022] Open
Abstract
Considerable data support a role for glycinergic ventromedial medulla neurons in the mediation of the postsynaptic inhibition of spinal motoneurons necessary for the motor atonia of rapid-eye movement (REM) sleep in cats. These data are, however, difficult to reconcile with the fact that large lesions of the rostral ventral medulla do not result in loss of REM atonia in rats. In the present study, we sought to clarify which medullary networks in rodents are responsible for REM motor atonia by retrogradely tracing inputs to the spinal ventral horn from the medulla, ablating these medullary sources to determine their effects on REM atonia and using transgenic mice to identify the neurotransmitter(s) involved. Our results reveal a restricted region within the ventromedial medulla, termed here the "supraolivary medulla" (SOM), which contains glutamatergic neurons that project to the spinal ventral horn. Cell-body specific lesions of the SOM resulted in an intermittent loss of muscle atonia, taking the form of exaggerated phasic muscle twitches, during REM sleep. A concomitant reduction in REM sleep time was observed in the SOM-lesioned animals. We next used mice with lox-P modified alleles of either the glutamate or GABA/glycine vesicular transporters to selectively eliminate glutamate or GABA/glycine neurotransmission from SOM neurons. Loss of SOM glutamate release, but not SOM GABA/glycine release, resulted in exaggerated muscle twitches during REM sleep that were similar to those observed after SOM lesions in rats. These findings, together, demonstrate that SOM glutamatergic neurons comprise key elements of the medullary circuitry mediating REM atonia.
Collapse
Affiliation(s)
| | | | - Qingchun Tong
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215
| | - Jun Lu
- Department of Neurology, Division of Sleep Medicine and
| |
Collapse
|
39
|
Fort P, Bassetti CL, Luppi PH. Alternating vigilance states: new insights regarding neuronal networks and mechanisms. Eur J Neurosci 2009; 29:1741-53. [DOI: 10.1111/j.1460-9568.2009.06722.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
40
|
Sapin E, Lapray D, Bérod A, Goutagny R, Léger L, Ravassard P, Clément O, Hanriot L, Fort P, Luppi PH. Localization of the brainstem GABAergic neurons controlling paradoxical (REM) sleep. PLoS One 2009; 4:e4272. [PMID: 19169414 PMCID: PMC2629845 DOI: 10.1371/journal.pone.0004272] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Accepted: 12/08/2008] [Indexed: 01/03/2023] Open
Abstract
Paradoxical sleep (PS) is a state characterized by cortical activation, rapid eye movements and muscle atonia. Fifty years after its discovery, the neuronal network responsible for the genesis of PS has been only partially identified. We recently proposed that GABAergic neurons would have a pivotal role in that network. To localize these GABAergic neurons, we combined immunohistochemical detection of Fos with non-radioactive in situ hybridization of GAD67 mRNA (GABA synthesis enzyme) in control rats, rats deprived of PS for 72 h and rats allowed to recover after such deprivation. Here we show that GABAergic neurons gating PS (PS-off neurons) are principally located in the ventrolateral periaqueductal gray (vlPAG) and the dorsal part of the deep mesencephalic reticular nucleus immediately ventral to it (dDpMe). Furthermore, iontophoretic application of muscimol for 20 min in this area in head-restrained rats induced a strong and significant increase in PS quantities compared to saline. In addition, we found a large number of GABAergic PS-on neurons in the vlPAG/dDPMe region and the medullary reticular nuclei known to generate muscle atonia during PS. Finally, we showed that PS-on neurons triggering PS localized in the SLD are not GABAergic. Altogether, our results indicate that multiple populations of PS-on GABAergic neurons are distributed in the brainstem while only one population of PS-off GABAergic neurons localized in the vlPAG/dDpMe region exist. From these results, we propose a revised model for PS control in which GABAergic PS-on and PS-off neurons localized in the vlPAG/dDPMe region play leading roles.
Collapse
Affiliation(s)
- Emilie Sapin
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Damien Lapray
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Anne Bérod
- CNRS, FRE3006, Pharmacologie et Imagerie de la neurotransmission sérotoninergique, Université Lyon1, Lyon, France
| | - Romain Goutagny
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Lucienne Léger
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Pascal Ravassard
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Olivier Clément
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Lucie Hanriot
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Patrice Fort
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
| | - Pierre-Hervé Luppi
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Lyon, France
- * E-mail:
| |
Collapse
|
41
|
Soja PJ. Glycine-mediated postsynaptic inhibition is responsible for REM sleep atonia. Sleep 2009; 31:1483-6. [PMID: 19014067 DOI: 10.1093/sleep/31.11.1483] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Peter J Soja
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver BC, Canada.
| |
Collapse
|
42
|
Taepavarapruk N, Taepavarapruk P, John J, Lai YY, Siegel JM, Phillips AG, McErlane SA, Soja PJ. State-dependent changes in glutamate, glycine, GABA, and dopamine levels in cat lumbar spinal cord. J Neurophysiol 2008; 100:598-608. [PMID: 18353913 DOI: 10.1152/jn.01231.2007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies have indicated that the glycine receptor antagonist strychnine and the gamma-aminobutyric acid type A (GABA A) receptor antagonist bicuculline reduced the rapid-eye-movement (REM) sleep-specific inhibition of sensory inflow via the dorsal spinocerebellar tract (DSCT). These findings imply that the spinal release of glycine and GABA may be due directly to the REM sleep-specific activation of reticulospinal neurons and/or glutamate-activated last-order spinal interneurons. This study used in vivo microdialysis and high-performance liquid chromatography analysis techniques to provide evidence for these possibilities. Microdialysis probes were stereotaxically positioned in the L3 spinal cord gray matter corresponding to sites where maximal cerebellar-evoked field potentials or individual DSCT and nearby spinoreticular tract (SRT) neurons could be recorded. Glutamate, glycine, and GABA levels significantly increased during REM sleep by approximately 48, 48, and 14%, respectively, compared with the control state of wakefulness. In contrast, dopamine levels significantly decreased by about 28% during REM sleep compared with wakefulness. During the state of wakefulness, electrical stimulation of the nucleus reticularis gigantocellularis (NRGc) at intensities sufficient to inhibit DSCT neuron activity, also significantly increased glutamate and glycine levels by about 69 and 45%, respectively, but not GABA or dopamine levels. We suggest that the reciprocal changes in the release of glutamate, glycine, and GABA versus dopamine during REM sleep contribute to the reduction of sensory inflow to higher brain centers via the DSCT and nearby SRT during this behavioral state. The neural pathways involved in this process likely include reticulo- and diencephalospinal and spinal interneurons.
Collapse
Affiliation(s)
- N Taepavarapruk
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, Canada, V6T 1Z3
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Shehab S, Alzigali L, Madathil M, Redgrave P. Pharmacological evidence for an anticonvulsant relay in the rat ventromedial medulla. Eur J Neurosci 2007; 26:2585-94. [DOI: 10.1111/j.1460-9568.2007.05851.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Luppi PH, Gervasoni D, Verret L, Goutagny R, Peyron C, Salvert D, Leger L, Fort P. Paradoxical (REM) sleep genesis: the switch from an aminergic-cholinergic to a GABAergic-glutamatergic hypothesis. ACTA ACUST UNITED AC 2007; 100:271-83. [PMID: 17689057 DOI: 10.1016/j.jphysparis.2007.05.006] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the middle of the last century, Michel Jouvet discovered paradoxical sleep (PS), a sleep phase paradoxically characterized by cortical activation and rapid eye movements and a muscle atonia. Soon after, he showed that it was still present in "pontine cats" in which all structures rostral to the brainstem have been removed. Later on, it was demonstrated that the pontine peri-locus coeruleus alpha (peri-LCalpha in cats, corresponding to the sublaterodorsal nucleus, SLD, in rats) is responsible for PS onset. It was then proposed that the onset and maintenance of PS is due to a reciprocal inhibitory interaction between neurons presumably cholinergic specifically active during PS localized in this region and monoaminergic neurons. In the last decade, we have tested this hypothesis with our model of head-restrained rats and functional neuroanatomical studies. Our results confirmed that the SLD in rats contains the neurons responsible for the onset and maintenance of PS. They further indicate that (1) these neurons are non-cholinergic possibly glutamatergic neurons, (2) they directly project to the glycinergic premotoneurons localized in the medullary ventral gigantocellular reticular nucleus (GiV), (3) the main neurotransmitter responsible for their inhibition during waking (W) and slow wave sleep (SWS) is GABA rather than monoamines, (4) they are constantly and tonically excited by glutamate and (5) the GABAergic neurons responsible for their tonic inhibition during W and SWS are localized in the deep mesencephalic reticular nucleus (DPMe). We also showed that the tonic inhibition of locus coeruleus (LC) noradrenergic and dorsal raphe (DRN) serotonergic neurons during sleep is due to a tonic GABAergic inhibition by neurons localized in the dorsal paragigantocellular reticular nucleus (DPGi) and the ventrolateral periaqueductal gray (vlPAG). We propose that these GABAergic neurons also inhibit the GABAergic neurons of the DPMe at the onset and during PS and are therefore responsible for the onset and maintenance of PS.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- UMR5167 CNRS, Faculté de Médecine Laennec, Institut Fédératif des Neurosciences de Lyon (IFR 19), Université Claude Bernard Lyon I, 7, Rue Guillaume Paradin, 69372 Lyon cedex 08, France.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hossaini M, French PJ, Holstege JC. Distribution of glycinergic neuronal somata in the rat spinal cord. Brain Res 2007; 1142:61-9. [PMID: 17336274 DOI: 10.1016/j.brainres.2007.01.078] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 01/02/2007] [Accepted: 01/12/2007] [Indexed: 01/09/2023]
Abstract
Glycine transporter 2 (GlyT2) mRNA is exclusively expressed in glycinergic neurons, and is presently considered a reliable marker for glycinergic neuronal somata. In this study, we have performed non-radioactive in situ hybridization to localize GlyT2 mRNA in fixed free-floating sections of cervical (C2 and C6), thoracic (T5), lumbar (L2 and L5) and sacral (S1) segments of the rat spinal cord. The results showed that in all segments the majority of the GlyT2 mRNA labeled (glycinergic) neuronal somata was present in the deep dorsal horn and the intermediate zone (laminae III-VIII), with around 50% (range 43.7-70.9%) in laminae VII&VIII. In contrast, the superficial dorsal horn, the motoneuronal cell groups and the area around the central canal contained only few glycinergic neuronal somata. The density (number of glycinergic neuronal somata per mm(2)) was also low in these areas, while the highest densities were found in laminae V to VIII. The lateral spinal nucleus and the lateral cervical nucleus also contained a limited number of glycinergic neurons. Our findings showed that the distribution pattern of the glycinergic neuronal somata is similar in all the examined segments. The few differences that were found in the relative laminar distribution between some of the segments, are most likely due to technical reasons. We therefore conclude that the observed distribution pattern of glycinergic neuronal somata is present throughout the spinal cord. Our findings further showed that the non-radioactive in situ hybridization technique for identifying GlyT2 mRNA in fixed free-floating sections is a highly efficient tool for identifying glycinergic neurons in the spinal cord.
Collapse
Affiliation(s)
- Mehdi Hossaini
- Department of Neuroscience, Erasmus MC, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | |
Collapse
|
46
|
Morales FR, Sampogna S, Rampon C, Luppi PH, Chase MH. Brainstem glycinergic neurons and their activation during active (rapid eye movement) sleep in the cat. Neuroscience 2006; 142:37-47. [PMID: 16891059 DOI: 10.1016/j.neuroscience.2006.05.066] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 05/19/2006] [Accepted: 05/23/2006] [Indexed: 10/24/2022]
Abstract
It is well established that, during rapid eye movement (REM) sleep, somatic motoneurons are subjected to a barrage of inhibitory synaptic potentials that are mediated by glycine. However, the source of this inhibition, which is crucial for the maintenance and preservation of REM sleep, has not been identified. Consequently, the present study was undertaken to determine in cats the location of the glycinergic neurons, that are activated during active sleep, and are responsible for the postsynaptic inhibition of motoneurons that occurs during this state. For this purpose, a pharmacologically-induced state of active sleep (AS-carbachol) was employed. Antibodies against glycine-conjugated proteins were used to identify glycinergic neurons and immunocytochemical techniques to label the Fos protein were employed to identify activated neurons. Two distinct populations of glycinergic neurons that expressed c-fos were distinguished. One population was situated within the nucleus reticularis gigantocellularis (NRGc) and nucleus magnocellularis (Mc) in the rostro-ventral medulla; this group of neurons extended caudally to the ventral portion of the nucleus paramedianus reticularis (nPR). Forty percent of the glycinergic neurons in the NRGc and Mc and 25% in the nPR expressed c-fos during AS-carbachol. A second population was located in the caudal medulla adjacent to the nucleus ambiguus (nAmb), wherein 40% of the glycinergic cells expressed c-fos during AS-carbachol. Neither population of glycinergic cells expressed c-fos during quiet wakefulness or quiet (non-rapid eye movement) sleep. We suggest that the population of glycinergic neurons in the NRGc, Mc, and nPR participates in the inhibition of somatic brainstem motoneurons during active sleep. These neurons may also be responsible for the inhibition of sensory and other processes during this state. It is likely that the group of glycinergic neurons adjacent to the nucleus ambiguus (nAmb) is responsible for the active sleep-selective inhibition of motoneurons that innervate the muscles of the larynx and pharynx.
Collapse
Affiliation(s)
- F R Morales
- WebSciences International, 1251 Westwood Boulevard, Los Angeles, CA 90024, USA
| | | | | | | | | |
Collapse
|
47
|
Shehab S, McGonigle D, Hughes DI, Todd AJ, Redgrave P. Anatomical evidence for an anticonvulsant relay in the rat ventromedial medulla. Eur J Neurosci 2005; 22:1431-44. [PMID: 16190897 DOI: 10.1111/j.1460-9568.2005.04326.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pharmacological manipulation of the ventrolateral pontine reticular formation (vlPRF) of rats has an anticonvulsant effect in the maximal electroshock model of epilepsy. This study presents three anatomical experiments that determine the efferent projections from this region likely to mediate this anticonvulsant effect. In the first, the anterograde tracer biotinylated dextran amine (BDA) was injected into the vlPRF. A strong projection to the ventromedial medullary reticular formation (vmMRF) was revealed which continued only weakly to the spinal cord. In the second experiment, double-label procedures were used to indicate whether the BDA-labelled terminals from the vlPRF make contacts with neurons in vmMRF, retrogradely labelled with cholera-toxin B subunit from the lumbar spinal cord. Sections of the vmMRF were examined by: (i) light microscopy which showed significant overlap between terminals from vlPRF and retrogradely-labelled reticulospinal cells; (ii) confocal microscopy which showed labelled terminals in close association with reticulospinal cell bodies; and (iii) electron microscopy which showed vlPRF terminals making synaptic contact with reticulospinal neurons. Finally, immunohistochemical procedures in combination with anterograde tracing revealed that significant numbers of terminals labelled from vlPRF were also positive for markers of glutamatergic or GABAergic neurotransmission. This suggests that the projection from the vlPRF to the vmMRF is likely to include several different functional components. These connections could represent a final critical link of an anticonvulsant circuit that originates in the dorsal midbrain and projects via relays in the vlPRF and the vmMRF to interact with the low-level motor circuitry in the spinal cord.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al-Ain, UAE.
| | | | | | | | | |
Collapse
|
48
|
Shigenaga Y, Moritani M, Oh SJ, Park KP, Paik SK, Bae JY, Kim HN, Ma SK, Park CW, Yoshida A, Ottersen OP, Bae YC. The distribution of inhibitory and excitatory synapses on single, reconstructed jaw-opening motoneurons in the cat. Neuroscience 2005; 133:507-18. [PMID: 15878646 DOI: 10.1016/j.neuroscience.2005.02.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Revised: 02/03/2005] [Accepted: 02/12/2005] [Indexed: 11/21/2022]
Abstract
In a previous study, we reported that the distribution of inhibitory input, in contrast to excitatory input, decreased somatofugally along dendrites of cat jaw-closing alpha-motoneurons [J Comp Neurol 414 (1999) 454]. The present study examined the distribution of GABA, glycine, and glutamate immunopositive boutons covering horseradish peroxidase-labeled cat jaw-opening motoneurons. The motoneurons were divided into four compartments: the soma, and primary, intermediate, and distal dendrites. Ninety-seven percent of the total number of studied boutons had immunoreactivity for at least one of the three amino acids. The proportion of boutons immunoreactive for GABA and/or glycine was lower than the proportion of boutons immunoreactive for glutamate. Boutons immunoreactive to glycine alone were more numerous than boutons double-labeled for GABA and glycine, which, in turn, occurred more frequently than boutons immunoreactive to GABA alone. The percentage synaptic covering (proportion of membrane covered by synaptic boutons) of the putatively excitatory (glutamate containing) and putatively inhibitory (GABA and/or glycine containing) boutons decreased somatofugally along the dendrites. Such systematic variations were not seen in the packing density (number of boutons per 100 microm(2)); the packing density showed a distinct drop between the soma and primary dendrites but did not differ significantly among the three dendritic compartments. Overall, the packing density was slightly higher for the putatively excitatory boutons than for the inhibitory ones. When taken together with previous analyses of jaw-closing alpha-motoneurons the present data on jaw-opening alpha-motoneurons indicate that the two types of neuron differ in regard to the nature of synaptic integration in the dendritic tree.
Collapse
Affiliation(s)
- Y Shigenaga
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Jung-Gu, Daegu 700-412, Korea.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Verret L, Léger L, Fort P, Luppi PH. Cholinergic and noncholinergic brainstem neurons expressing Fos after paradoxical (REM) sleep deprivation and recovery. Eur J Neurosci 2005; 21:2488-504. [PMID: 15932606 DOI: 10.1111/j.1460-9568.2005.04060.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is well accepted that populations of neurons responsible for the onset and maintenance of paradoxical sleep (PS) are restricted to the brainstem. To localize the structures involved and to reexamine the role of mesopontine cholinergic neurons, we compared the distribution of Fos- and choline acetyltransferase-labelled neurons in the brainstem of control rats, rats selectively deprived of PS for approximately 72 h and rats allowed to recover from such deprivation. Only a few cholinergic neurons from the laterodorsal (LDTg) and pedunculopontine tegmental nuclei were Fos-labelled after PS recovery. In contrast, a large number of noncholinergic Fos-labelled cells positively correlated with the percentage of time spent in PS was observed in the LDTg, sublaterodorsal, alpha and ventral gigantocellular reticular nuclei, structures known to contain neurons specifically active during PS. In addition, a large number of Fos-labelled cells were seen after PS rebound in the lateral, ventrolateral and dorsal periaqueductal grey, dorsal and lateral paragigantocellular reticular nuclei and the nucleus raphe obscurus. Interestingly, half of the cells in the latter nucleus were immunoreactive to choline acetyltransferase. In contrast to the well-accepted hypothesis, our results strongly suggest that neurons active during PS, recorded in the mesopontine cholinergic nuclei, are in the great majority noncholinergic. Our findings further demonstrate that many brainstem structures not previously identified as containing neurons active during PS contain cholinergic or noncholinergic neurons active during PS, and these structures may therefore play a key role during this state. Altogether, our results open a new avenue of research to identify the specific role of the populations of neurons revealed, their interrelations and their neurochemical identity.
Collapse
Affiliation(s)
- Laure Verret
- CNRS UMR 5167, Institut Fédératif des Neurosciences de Lyon (IFR 19), Faculté de médecine RTH Laennec, 7, rue Guillaume Paradin, 69372 Lyon Cedex 08, France
| | | | | | | |
Collapse
|
50
|
Pagnotta SE, Lape R, Quitadamo C, Nistri A. Pre- and postsynaptic modulation of glycinergic and gabaergic transmission by muscarinic receptors on rat hypoglossal motoneurons in vitro. Neuroscience 2005; 130:783-95. [PMID: 15590160 DOI: 10.1016/j.neuroscience.2004.09.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2004] [Indexed: 11/16/2022]
Abstract
The motor output of hypoglossal motoneurons to tongue muscles takes place in concert with the respiratory rhythm and is determined by the balance between excitatory glutamatergic transmission and inhibitory transmission mediated by glycine or GABA. The relative contribution by these transmitters is a phasic phenomenon modulated by other transmitters. We examined how metabotropic muscarinic receptors, widely expressed in the brainstem where they excite cranial motor nuclei, might influence synaptic activity mediated by GABA or glycine. For this purpose, using thin slices of the neonatal rat brainstem, we recorded (under whole-cell patch clamp) glycinergic or GABAergic responses from visually identified hypoglossal motoneurons after pharmacological block of glutamatergic transmission. Muscarine inhibited spontaneous and electrically induced events mediated by GABA or glycine. The amplitude of glycinergic miniature inhibitory postsynaptic currents was slightly reduced by muscarine, while GABAergic miniature inhibitory postsynaptic currents were unaffected. Motoneuron currents induced by focally applied GABA and glycine were depressed by muscarine with stronger reduction in glycine-mediated responses. Histochemical observations indicated the presence of M1, M2 and M5 subtypes of muscarinic receptors in the neonatal hypoglossal nucleus. These results suggest that muscarine potently depressed inhibitory neurotransmission on brainstem motoneurons, and that this action was exerted via preterminal and extrasynaptic receptors. Since the large reduction in inhibitory neurotransmission may contribute to overall excitation of brainstem motoneurons by muscarinic receptors, these data might help to understand the central components of action of antimuscarinic agents in preanesthetic medication or against motion sickness.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Data Interpretation, Statistical
- Electric Stimulation
- Electrophysiology
- Excitatory Postsynaptic Potentials
- Glycine/physiology
- Hypoglossal Nerve/drug effects
- Hypoglossal Nerve/physiology
- Immunohistochemistry
- In Vitro Techniques
- Motor Neurons/drug effects
- Motor Neurons/physiology
- Muscarine/pharmacology
- Muscarinic Agonists/pharmacology
- Patch-Clamp Techniques
- Rats
- Rats, Wistar
- Receptors, GABA/drug effects
- Receptors, GABA/physiology
- Receptors, Glycine/drug effects
- Receptors, Glycine/physiology
- Receptors, Muscarinic/drug effects
- Receptors, Muscarinic/physiology
- Receptors, Presynaptic/drug effects
- Receptors, Presynaptic/physiology
- Synapses/drug effects
- Synapses/physiology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- gamma-Aminobutyric Acid/physiology
Collapse
Affiliation(s)
- S E Pagnotta
- Neurobiology Sector and INFM Unit, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | | | | | | |
Collapse
|