1
|
Setzer B, Fultz NE, Gomez DEP, Williams SD, Bonmassar G, Polimeni JR, Lewis LD. A temporal sequence of thalamic activity unfolds at transitions in behavioral arousal state. Nat Commun 2022; 13:5442. [PMID: 36114170 PMCID: PMC9481532 DOI: 10.1038/s41467-022-33010-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Awakening from sleep reflects a profound transformation in neural activity and behavior. The thalamus is a key controller of arousal state, but whether its diverse nuclei exhibit coordinated or distinct activity at transitions in behavioral arousal state is unknown. Using fast fMRI at ultra-high field (7 Tesla), we measured sub-second activity across thalamocortical networks and within nine thalamic nuclei to delineate these dynamics during spontaneous transitions in behavioral arousal state. We discovered a stereotyped sequence of activity across thalamic nuclei and cingulate cortex that preceded behavioral arousal after a period of inactivity, followed by widespread deactivation. These thalamic dynamics were linked to whether participants subsequently fell back into unresponsiveness, with unified thalamic activation reflecting maintenance of behavior. These results provide an outline of the complex interactions across thalamocortical circuits that orchestrate behavioral arousal state transitions, and additionally, demonstrate that fast fMRI can resolve sub-second subcortical dynamics in the human brain.
Collapse
Affiliation(s)
- Beverly Setzer
- Graduate Program for Neuroscience, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Nina E Fultz
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Daniel E P Gomez
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Giorgio Bonmassar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura D Lewis
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA.
| |
Collapse
|
2
|
Abstract
Brain PCO2 is sensed primarily via changes in [H+]. Small pH changes are detected in the medulla oblongata and trigger breathing adjustments that help maintain arterial PCO2 constant. Larger perturbations of brain CO2/H+, possibly also sensed elsewhere in the CNS, elicit arousal, dyspnea, and stress, and cause additional breathing modifications. The retrotrapezoid nucleus (RTN), a rostral medullary cluster of glutamatergic neurons identified by coexpression of Phoxb and Nmb transcripts, is the lynchpin of the central respiratory chemoreflex. RTN regulates breathing frequency, inspiratory amplitude, and active expiration. It is exquisitely responsive to acidosis in vivo and maintains breathing autorhythmicity during quiet waking, slow-wave sleep, and anesthesia. The RTN response to [H+] is partly an intrinsic neuronal property mediated by proton sensors TASK-2 and GPR4 and partly a paracrine effect mediated by astrocytes and the vasculature. The RTN also receives myriad excitatory or inhibitory synaptic inputs including from [H+]-responsive neurons (e.g., serotonergic). RTN is silenced by moderate hypoxia. RTN inactivity (periodic or sustained) contributes to periodic breathing and, likely, to central sleep apnea. RTN development relies on transcription factors Egr2, Phox2b, Lbx1, and Atoh1. PHOX2B mutations cause congenital central hypoventilation syndrome; they impair RTN development and consequently the central respiratory chemoreflex.
Collapse
Affiliation(s)
- Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States.
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
3
|
Masneuf S, Imbach LL, Büchele F, Colacicco G, Penner M, Moreira CG, Ineichen C, Jahanshahi A, Temel Y, Baumann CR, Noain D. Altered sleep intensity upon DBS to hypothalamic sleep-wake centers in rats. Transl Neurosci 2021; 12:611-625. [PMID: 35070444 PMCID: PMC8729228 DOI: 10.1515/tnsci-2020-0202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 11/15/2022] Open
Abstract
Deep brain stimulation (DBS) has been scarcely investigated in the field of sleep research. We hypothesize that DBS onto hypothalamic sleep- and wake-promoting centers will produce significant neuromodulatory effects and potentially become a therapeutic strategy for patients suffering severe, drug-refractory sleep–wake disturbances. We aimed to investigate whether continuous electrical high-frequency DBS, such as that often implemented in clinical practice, in the ventrolateral preoptic nucleus (VLPO) or the perifornical area of the posterior lateral hypothalamus (PeFLH), significantly modulates sleep–wake characteristics and behavior. We implanted healthy rats with electroencephalographic/electromyographic electrodes and recorded vigilance states in parallel to bilateral bipolar stimulation of VLPO and PeFLH at 125 Hz and 90 µA over 24 h to test the modulating effects of DBS on sleep–wake proportions, stability and spectral power in relation to the baseline. We unexpectedly found that VLPO DBS at 125 Hz deepens slow-wave sleep (SWS) as measured by increased delta power, while sleep proportions and fragmentation remain unaffected. Thus, the intensity, but not the amount of sleep or its stability, is modulated. Similarly, the proportion and stability of vigilance states remained altogether unaltered upon PeFLH DBS but, in contrast to VLPO, 125 Hz stimulation unexpectedly weakened SWS, as evidenced by reduced delta power. This study provides novel insights into non-acute functional outputs of major sleep–wake centers in the rat brain in response to electrical high-frequency stimulation, a paradigm frequently used in human DBS. In the conditions assayed, while exerting no major effects on the sleep–wake architecture, hypothalamic high-frequency stimulation arises as a provocative sleep intensity-modulating approach.
Collapse
Affiliation(s)
- Sophie Masneuf
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas L Imbach
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Fabian Büchele
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Marco Penner
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carlos G Moreira
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Ineichen
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, DPPP, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Ali Jahanshahi
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.,Center of Competence Sleep & Health, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [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
|
5
|
Agostinelli LJ, Geerling JC, Scammell TE. Basal forebrain subcortical projections. Brain Struct Funct 2019; 224:1097-1117. [PMID: 30612231 PMCID: PMC6500474 DOI: 10.1007/s00429-018-01820-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/16/2018] [Indexed: 12/25/2022]
Abstract
The basal forebrain (BF) contains at least three distinct populations of neurons (cholinergic, glutamatergic, and GABA-ergic) across its different regions (medial septum, diagonal band, magnocellular preoptic area, and substantia innominata). Much attention has focused on the BF's ascending projections to cortex, but less is known about descending projections to subcortical regions. Given the neurochemical and anatomical heterogeneity of the BF, we used conditional anterograde tracing to map the patterns of subcortical projections from multiple BF regions and neurochemical cell types using mice that express Cre recombinase only in cholinergic, glutamatergic, or GABAergic neurons. We confirmed that different BF regions innervate distinct subcortical targets, with more subcortical projections arising from neurons in the caudal and lateral BF (substantia innominata and magnocellular preoptic area). Additionally, glutamatergic and GABAergic BF neurons have distinct patterns of descending projections, while cholinergic descending projections are sparse. Considering the intensity of glutamatergic and GABAergic descending projections, the BF likely acts through subcortical targets to promote arousal, motivation, and other behaviors.
Collapse
Affiliation(s)
- Lindsay J Agostinelli
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Joel C Geerling
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Gobbi G, Comai S. Differential Function of Melatonin MT 1 and MT 2 Receptors in REM and NREM Sleep. Front Endocrinol (Lausanne) 2019; 10:87. [PMID: 30881340 PMCID: PMC6407453 DOI: 10.3389/fendo.2019.00087] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 11/13/2022] Open
Abstract
The pathophysiological function of the G-protein coupled melatonin MT1 and MT2 receptors has not yet been well-clarified. Recent advancements using selective MT1/ MT2 receptor ligands and MT1/MT2 receptor knockout mice have suggested that the activation of the MT1 receptors are mainly implicated in the regulation of rapid eye movement (REM) sleep, whereas the MT2 receptors selectively increase non-REM (NREM) sleep. Studies in mutant mice show that MT1 knockout mice have an increase in NREM sleep and a decrease in REM sleep, while MT2 knockout mice a decrease in NREM sleep. The localization of MT1 receptors is also distinct from MT2 receptors; for example, MT2 receptors are located in the reticular thalamus (NREM area), while the MT1 receptors in the Locus Coeruleus and lateral hypothalamus (REM areas). Altogether, these findings suggest that these two receptors not only have a very specialized function in sleep, but that they may also modulate opposing effects. These data also suggest that mixed MT1-MT2 receptors ligands are not clinically recommended given their opposite roles in physiological functions, confirmed by the modest effects of melatonin or MT1/MT2 non-selective agonists when used in both preclinical and clinical studies as hypnotic drugs. In sum, MT1 and MT2 receptors have specific roles in the modulation of sleep, and consequently, selective ligands with agonist, antagonist, or partial agonist properties could have therapeutic potential for sleep; while the MT2 agonists or partial agonists might be indicated for NREM-related sleep and/or anxiety disorders, the MT1 agonists or partial agonists might be so for REM-related sleep disorders. Furthermore, MT1 but not MT2 receptors seem involved in the regulation of the circadian rhythm. Future research will help further develop MT1 and/or MT2 receptors as targets for neuropsychopharmacology drug development.
Collapse
Affiliation(s)
- Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada
- San Raffaele Scientific Institute and Vita Salute University, Milan, Italy
| |
Collapse
|
7
|
Hypothalamic Neurons that Regulate Feeding Can Influence Sleep/Wake States Based on Homeostatic Need. Curr Biol 2018; 28:3736-3747.e3. [PMID: 30471995 DOI: 10.1016/j.cub.2018.09.055] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Abstract
Eating and sleeping represent two mutually exclusive behaviors that satisfy distinct homeostatic needs. Because an animal cannot eat and sleep at the same time, brain systems that regulate energy homeostasis are likely to influence sleep/wake behavior. Indeed, previous studies indicate that animals adjust sleep cycles around periods of food need and availability. Furthermore, hormones that affect energy homeostasis also affect sleep/wake states: the orexigenic hormone ghrelin promotes wakefulness, and the anorexigenic hormones leptin and insulin increase the duration of slow-wave sleep. However, whether neural populations that regulate feeding can influence sleep/wake states is unknown. The hypothalamic arcuate nucleus contains two neuronal populations that exert opposing effects on energy homeostasis: agouti-related protein (AgRP)-expressing neurons detect caloric need and orchestrate food-seeking behavior, whereas activity in pro-opiomelanocortin (POMC)-expressing neurons induces satiety. We tested the hypotheses that AgRP neurons affect sleep homeostasis by promoting states of wakefulness, whereas POMC neurons promote states of sleep. Indeed, optogenetic or chemogenetic stimulation of AgRP neurons in mice promoted wakefulness while decreasing the quantity and integrity of sleep. Inhibition of AgRP neurons rescued sleep integrity in food-deprived mice, highlighting the physiological importance of AgRP neuron activity for the suppression of sleep by hunger. Conversely, stimulation of POMC neurons promoted sleep states and decreased sleep fragmentation in food-deprived mice. Interestingly, we also found that sleep deprivation attenuated the effects of AgRP neuron activity on food intake and wakefulness. These results indicate that homeostatic feeding neurons can hierarchically affect behavioral outcomes, depending on homeostatic need.
Collapse
|
8
|
Atkin T, Comai S, Gobbi G. Drugs for Insomnia beyond Benzodiazepines: Pharmacology, Clinical Applications, and Discovery. Pharmacol Rev 2018; 70:197-245. [PMID: 29487083 DOI: 10.1124/pr.117.014381] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although the GABAergic benzodiazepines (BZDs) and Z-drugs (zolpidem, zopiclone, and zaleplon) are FDA-approved for insomnia disorders with a strong evidence base, they have many side effects, including cognitive impairment, tolerance, rebound insomnia upon discontinuation, car accidents/falls, abuse, and dependence liability. Consequently, the clinical use of off-label drugs and novel drugs that do not target the GABAergic system is increasing. The purpose of this review is to analyze the neurobiological and clinical evidence of pharmacological treatments of insomnia, excluding the BZDs and Z-drugs. We analyzed the melatonergic agonist drugs, agomelatine, prolonged-release melatonin, ramelteon, and tasimelteon; the dual orexin receptor antagonist suvorexant; the modulators of the α2δ subunit of voltage-sensitive calcium channels, gabapentin and pregabalin; the H1 antagonist, low-dose doxepin; and the histamine and serotonin receptor antagonists, amitriptyline, mirtazapine, trazodone, olanzapine, and quetiapine. The pharmacology and mechanism of action of these treatments and the evidence-base for the use of these drugs in clinical practice is outlined along with novel pipelines. There is evidence to recommend suvorexant and low-dose doxepin for sleep maintenance insomnia; there is also sufficient evidence to recommend ramelteon for sleep onset insomnia. Although there is limited evidence for the use of the quetiapine, trazodone, mirtazapine, amitriptyline, pregabalin, gabapentin, agomelatine, and olanzapine as treatments for insomnia disorder, these drugs may improve sleep while successfully treating comorbid disorders, with a different side effect profile than the BZDs and Z-drugs. The unique mechanism of action of each drug allows for a more personalized and targeted medical management of insomnia.
Collapse
Affiliation(s)
- Tobias Atkin
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| |
Collapse
|
9
|
Tyree SM, de Lecea L. Lateral Hypothalamic Control of the Ventral Tegmental Area: Reward Evaluation and the Driving of Motivated Behavior. Front Syst Neurosci 2017; 11:50. [PMID: 28729827 PMCID: PMC5498520 DOI: 10.3389/fnsys.2017.00050] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/22/2017] [Indexed: 12/25/2022] Open
Abstract
The lateral hypothalamus (LH) plays an important role in many motivated behaviors, sleep-wake states, food intake, drug-seeking, energy balance, etc. It is also home to a heterogeneous population of neurons that express and co-express multiple neuropeptides including hypocretin (Hcrt), melanin-concentrating hormone (MCH), cocaine- and amphetamine-regulated transcript (CART) and neurotensin (NT). These neurons project widely throughout the brain to areas such as the locus coeruleus, the bed nucleus of the stria terminalis, the amygdala and the ventral tegmental area (VTA). Lateral hypothalamic projections to the VTA are believed to be important for driving behavior due to the involvement of dopaminergic reward circuitry. The purpose of this article is to review current knowledge regarding the lateral hypothalamic connections to the VTA and the role they play in driving these behaviors.
Collapse
Affiliation(s)
- Susan M Tyree
- Department of Psychiatry and Behavioral Sciences, Stanford UniversityStanford, CA, United States
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford UniversityStanford, CA, United States
| |
Collapse
|
10
|
Kong XP, Wang C, Xie JF, Zhao P, Dai LR, Shao YF, Lin JS, Hou YP. Neuropeptide S reduces propofol- or ketamine-induced slow wave states through activation of cognate receptors in the rat. Neuropeptides 2017; 63:59-66. [PMID: 28228241 DOI: 10.1016/j.npep.2017.02.081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/12/2017] [Accepted: 02/13/2017] [Indexed: 11/21/2022]
Abstract
Intracerebroventricular injection of NPS reduces the duration of the ketamine- or thiopental-induced loss of the righting reflex in rats. But the specific EEG activities are unknown. We therefore sought to examine the effects of the NPS-NPSR system on anesthetic-induced characteristics of EEG power spectra and sleep-wake profiles. NPS alone or together with an NPSR antagonist was injected intracerebroventricularly, whereas the propofol (50mg/kg) or ketamine (100mg/kg) was administrated intraperitoneally. NPS (1 or 2nmol) significantly reduced the amount of propofol-induced EEG delta activity and slow wave states (SWS). NPS (1 or 5nmol) significantly reduced the amount of ketamine-induced SWS and EEG delta activity. Cortical EEG power spectral analysis showed that, in saline-pretreated rats, propofol induced a marked increase in delta (0.5-4Hz) activity, decrease in theta (4.5-8.5Hz) activity, and decrease in high frequency activity (14.5-60Hz), while, in rats pretreated with 1nmol of NPS, the duration of delta activity was reduced, while its spectral pattern was not changed. Whereas injection of ketamine into saline-pretreated rats induced a marked increase in delta (0.5-4Hz) activity, a moderate increase in theta (4.5-8.5Hz) activity, and a marked decrease in high frequency (14.5-60Hz) activity. However, delta activity was reduced while theta activity increased under pretreatment with 1nmol of NPS. The inhibitory effect of NPS on anesthetic-induced SWS was characterized by a reduced SWS episode duration with no significant change in either episode number or latency to SWS. [D-Val5]NPS, an NPSR antagonist (20nmol), significantly attenuated the arousal-promoting effect of 1nmol of NPS, but had no effect on SWS when injected alone. We speculate that NPS significantly reduces anesthetic-induced SWS and EEG slow activity by selective activation of the NPSR, which, in turn, would trigger subsequent arousal pathways.
Collapse
Affiliation(s)
- Xiang-Pan Kong
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China; Department of Human Anatomy, Hunan Normal University, Changsha, China
| | - Can Wang
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| | - Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| | - Peng Zhao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| | - Li-Rong Dai
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| | - Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, University Claude Bernard Lyon 1, Lyon, France.
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Agostinelli LJ, Ferrari LL, Mahoney CE, Mochizuki T, Lowell BB, Arrigoni E, Scammell TE. Descending projections from the basal forebrain to the orexin neurons in mice. J Comp Neurol 2017; 525:1668-1684. [PMID: 27997037 PMCID: PMC5806522 DOI: 10.1002/cne.24158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 12/23/2022]
Abstract
The orexin (hypocretin) neurons play an essential role in promoting arousal, and loss of the orexin neurons results in narcolepsy, a condition characterized by chronic sleepiness and cataplexy. The orexin neurons excite wake-promoting neurons in the basal forebrain (BF), and a reciprocal projection from the BF back to the orexin neurons may help promote arousal and motivation. The BF contains at least three different cell types (cholinergic, glutamatergic, and γ-aminobutyric acid (GABA)ergic neurons) across its different regions (medial septum, diagonal band, magnocellular preoptic area, and substantia innominata). Given the neurochemical and anatomical heterogeneity of the BF, we mapped the pattern of BF projections to the orexin neurons across multiple BF regions and neuronal types. We performed conditional anterograde tracing using mice that express Cre recombinase only in neurons producing acetylcholine, glutamate, or GABA. We found that the orexin neurons are heavily apposed by axon terminals of glutamatergic and GABAergic neurons of the substantia innominata (SI) and magnocellular preoptic area, but there was no innervation by the cholinergic neurons. Channelrhodopsin-assisted circuit mapping (CRACM) demonstrated that glutamatergic SI neurons frequently form functional synapses with the orexin neurons, but, surprisingly, functional synapses from SI GABAergic neurons were rare. Considering their strong reciprocal connections, BF and orexin neurons likely work in concert to promote arousal, motivation, and other behaviors. J. Comp. Neurol. 525:1668-1684, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lindsay J Agostinelli
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Loris L Ferrari
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Takatoshi Mochizuki
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Bradford B Lowell
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Eban-Rothschild A, de Lecea L. Neuronal substrates for initiation, maintenance, and structural organization of sleep/wake states. F1000Res 2017; 6:212. [PMID: 28357049 PMCID: PMC5345773 DOI: 10.12688/f1000research.9677.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2017] [Indexed: 11/20/2022] Open
Abstract
Animals continuously alternate between sleep and wake states throughout their life. The daily organization of sleep and wakefulness is orchestrated by circadian, homeostatic, and motivational processes. Over the last decades, much progress has been made toward determining the neuronal populations involved in sleep/wake regulation. Here, we will discuss how the application of advanced
in vivo tools for cell type–specific manipulations now permits the functional interrogation of different features of sleep/wake state regulation: initiation, maintenance, and structural organization. We will specifically focus on recent studies examining the roles of wake-promoting neuronal populations.
Collapse
Affiliation(s)
- Ada Eban-Rothschild
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
13
|
The Neuropeptide Orexin-A Inhibits the GABAA Receptor by PKC and Ca2+/CaMKII-Dependent Phosphorylation of Its β1 Subunit. J Mol Neurosci 2017; 61:459-467. [DOI: 10.1007/s12031-017-0886-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
|
14
|
Optogenetic control of human neurons in organotypic brain cultures. Sci Rep 2016; 6:24818. [PMID: 27098488 PMCID: PMC4838935 DOI: 10.1038/srep24818] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/06/2016] [Indexed: 01/16/2023] Open
Abstract
Optogenetics is one of the most powerful tools in neuroscience, allowing for selective control of specific neuronal populations in the brain of experimental animals, including mammals. We report, for the first time, the application of optogenetic tools to human brain tissue providing a proof-of-concept for the use of optogenetics in neuromodulation of human cortical and hippocampal neurons as a possible tool to explore network mechanisms and develop future therapeutic strategies.
Collapse
|
15
|
Song C, Knöpfel T. Optogenetics enlightens neuroscience drug discovery. Nat Rev Drug Discov 2015; 15:97-109. [DOI: 10.1038/nrd.2015.15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
16
|
Castro DC, Cole SL, Berridge KC. Lateral hypothalamus, nucleus accumbens, and ventral pallidum roles in eating and hunger: interactions between homeostatic and reward circuitry. Front Syst Neurosci 2015; 9:90. [PMID: 26124708 PMCID: PMC4466441 DOI: 10.3389/fnsys.2015.00090] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 05/29/2015] [Indexed: 12/16/2022] Open
Abstract
The study of the neural bases of eating behavior, hunger, and reward has consistently implicated the lateral hypothalamus (LH) and its interactions with mesocorticolimbic circuitry, such as mesolimbic dopamine projections to nucleus accumbens (NAc) and ventral pallidum (VP), in controlling motivation to eat. The NAc and VP play special roles in mediating the hedonic impact (“liking”) and motivational incentive salience (“wanting”) of food rewards, and their interactions with LH help permit regulatory hunger/satiety modulation of food motivation and reward. Here, we review some progress that has been made regarding this circuitry and its functions: the identification of localized anatomical hedonic hotspots within NAc and VP for enhancing hedonic impact; interactions of NAc/VP hedonic hotspots with specific LH signals such as orexin; an anterior-posterior gradient of sites in NAc shell for producing intense appetitive eating vs. intense fearful reactions; and anatomically distributed appetitive functions of dopamine and mu opioid signals in NAc shell and related structures. Such findings help improve our understanding of NAc, VP, and LH interactions in mediating affective and motivation functions, including “liking” and “wanting” for food rewards.
Collapse
Affiliation(s)
- Daniel C Castro
- Department of Psychology, University of Michigan Ann Arbor, MI, USA
| | - Shannon L Cole
- Department of Psychology, University of Michigan Ann Arbor, MI, USA
| | - Kent C Berridge
- Department of Psychology, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
17
|
Kosse C, Burdakov D. A unifying computational framework for stability and flexibility of arousal. Front Syst Neurosci 2014; 8:192. [PMID: 25368557 PMCID: PMC4202806 DOI: 10.3389/fnsys.2014.00192] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/18/2014] [Indexed: 02/02/2023] Open
Abstract
Arousal and consciousness flexibly adjust to salient cues, but remain stable despite noise and disturbance. Diverse, highly interconnected neural networks govern the underlying transitions of behavioral state; these networks are robust but very complex. Frameworks from systems engineering provide powerful tools for understanding functional logic behind component complexity. From a general systems viewpoint, a minimum of three communicating control modules may enable flexibility and stability to coexist. Comparators would subtract current arousal from desired arousal, producing an error signal. Regulators would compute control signals from this error. Generators would convert control signals into arousal, which is fed back to comparators, to make the system noise-proof through self-correction. Can specific neurons correspond to these control elements? To explore this, here we consider the brain-wide orexin/hypocretin network, which is experimentally established to be vital for flexible and stable arousal. We discuss whether orexin neurons may act as comparators, and their target neurons as regulators and generators. Experiments are proposed for testing such predictions, based on computational simulations showing that comparators, regulators, and generators have distinct temporal signatures of activity. If some regulators integrate orexin-communicated errors, robust arousal control may be achieved via integral feedback (a basic engineering strategy for tracking a set-point despite noise). An integral feedback view also suggests functional roles for specific molecular aspects, such as differing life-spans of orexin peptides. The proposed framework offers a unifying logic for molecular, cellular, and network details of arousal systems, and provides insight into behavioral state transitions, complex behavior, and bases for disease.
Collapse
Affiliation(s)
- Christin Kosse
- Neurophysiology, MRC National Institute for Medical Research London, UK
| | - Denis Burdakov
- Neurophysiology, MRC National Institute for Medical Research London, UK ; MRC Centre for Developmental Neurobiology, King's College London London, UK
| |
Collapse
|
18
|
Narcolepsy patients have antibodies that stain distinct cell populations in rat brain and influence sleep patterns. Proc Natl Acad Sci U S A 2014; 111:E3735-44. [PMID: 25136085 DOI: 10.1073/pnas.1412189111] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Narcolepsy is a chronic sleep disorder, likely with an autoimmune component. During 2009 and 2010, a link between A(H1N1)pdm09 Pandemrix vaccination and onset of narcolepsy was suggested in Scandinavia. In this study, we searched for autoantibodies related to narcolepsy using a neuroanatomical array: rat brain sections were processed for immunohistochemistry/double labeling using patient sera/cerebrospinal fluid as primary antibodies. Sera from 89 narcoleptic patients, 52 patients with other sleep-related disorders (OSRDs), and 137 healthy controls were examined. Three distinct patterns of immunoreactivity were of particular interest: pattern A, hypothalamic melanin-concentrating hormone and proopiomelanocortin but not hypocretin/orexin neurons; pattern B, GABAergic cortical interneurons; and pattern C, mainly globus pallidus neurons. Altogether, 24 of 89 (27%) narcoleptics exhibited pattern A or B or C. None of the patterns were exclusive for narcolepsy but were also detected in the OSRD group at significantly lower numbers. Also, some healthy controls exhibited these patterns. The antigen of pattern A autoantibodies was identified as the common C-terminal epitope of neuropeptide glutamic acid-isoleucine/α-melanocyte-stimulating hormone (NEI/αMSH) peptides. Passive transfer experiments on rat showed significant effects of pattern A human IgGs on rapid eye movement and slow-wave sleep time parameters in the inactive phase and EEG θ-power in the active phase. We suggest that NEI/αMSH autoantibodies may interfere with the fine regulation of sleep, contributing to the complex pathogenesis of narcolepsy and OSRDs. Also, patterns B and C are potentially interesting, because recent data suggest a relevance of those brain regions/neuron populations in the regulation of sleep/arousal.
Collapse
|
19
|
What optogenetic stimulation is telling us (and failing to tell us) about fast neurotransmitters and neuromodulators in brain circuits for wake-sleep regulation. Curr Opin Neurobiol 2014; 29:165-71. [PMID: 25064179 DOI: 10.1016/j.conb.2014.07.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/07/2014] [Accepted: 07/07/2014] [Indexed: 12/19/2022]
Abstract
In the last eight years optogenetic tools have been widely used to identify functional synaptic connectivity between specific neuronal populations. Most of our knowledge comes from the photo-activation of channelrhodopsin-2 (ChR2) expressing inputs that release glutamate and GABA. More recent studies have been reporting releases of acetylcholine and biogenic amines but direct evidence for photo-evoked released of neuropeptides is still limited particularly in brain slice studies. The high fidelity in the responses with photo-evoked amino-acid transmission is ideal for ChR2-assisted circuit mapping and this approach has been successfully used in different fields of neuroscience. Conversely, neuropeptides employ a slow mode of communication and might require higher frequency and prolonged stimulations to be released. These factors may have contributed to the apparent lack of success for optogenetic release of neuropeptides. In addition, once released, neuropeptides often act on multiple sites and at various distances from the site of release resulting in a greater complexity of postsynaptic responses. Here, we focus on what optogenetics is telling us-and failing to tell us-about fast neurotransmitters and neuropeptides.
Collapse
|
20
|
Abstract
In 1998, our group discovered a cDNA that encoded the precursor of two putative neuropeptides that we called hypocretins for their hypothalamic expression and their similarity to the secretin family of neuropeptides. In the last 16 years, numerous studies have placed the hypocretin system as an integrator of homeostatic functions with a crucial, non-redundant function as arousal stabilizer. We recently applied optogenetic methods to interrogate the role of individual neuronal circuits in sleep-to-wake transitions. The neuronal connections between the hypocretin system and the locus coeruleus (LC) seem to be crucial in establishing the appropriate dynamic of spontaneous awakenings.
Collapse
|
21
|
Orexin/hypocretin system modulates amygdala-dependent threat learning through the locus coeruleus. Proc Natl Acad Sci U S A 2013; 110:20260-5. [PMID: 24277819 DOI: 10.1073/pnas.1320325110] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Survival in a dangerous environment requires learning about stimuli that predict harm. Although recent work has focused on the amygdala as the locus of aversive memory formation, the hypothalamus has long been implicated in emotional regulation, and the hypothalamic neuropeptide orexin (hypocretin) is involved in anxiety states and arousal. Nevertheless, little is known about the role of orexin in aversive memory formation. Using a combination of behavioral pharmacology, slice physiology, and optogenetic techniques, we show that orexin acts upstream of the amygdala via the noradrenergic locus coeruleus to enable threat (fear) learning, specifically during the aversive event. Our results are consistent with clinical studies linking orexin levels to aversive learning and anxiety in humans and dysregulation of the orexin system may contribute to the etiology of fear and anxiety disorders.
Collapse
|
22
|
Hastings MH, Goedert M. Circadian clocks and neurodegenerative diseases: time to aggregate? Curr Opin Neurobiol 2013; 23:880-7. [PMID: 23797088 PMCID: PMC3782660 DOI: 10.1016/j.conb.2013.05.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 05/14/2013] [Accepted: 05/22/2013] [Indexed: 12/15/2022]
Abstract
The major neurodegenerative diseases are characterised by a disabling loss of the daily pattern of sleep and wakefulness, which may be reflective of a compromise to the underlying circadian clock that times the sleep cycle. At a molecular level, the canonical property of neurodegenerative diseases is aberrant aggregation of otherwise soluble neuronal proteins. They can thus be viewed as disturbances of proteostasis, raising the question whether the two features - altered daily rhythms and molecular aggregation - are related. Recent discoveries have highlighted the fundamental contribution of circadian clocks to the correct ordering of daily cellular metabolic cycles, imposing on peripheral organs such as the liver a strict programme that alternates between anabolic and catabolic states. The discovery that circadian mechanisms are active in local brain regions suggests that they may impinge upon physiological and pathological elements that influence pro-neurodegenerative aggregation. This review explores how introducing the dimension of circadian time and the circadian clock might refine the analysis of aberrant aggregation, thus expanding our perspective on the cell biology common to neurodegenerative diseases.
Collapse
Affiliation(s)
- Michael H Hastings
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| | | |
Collapse
|
23
|
Beck P, Urbano FJ, Williams DK, Garcia-Rill E. Effects of leptin on pedunculopontine nucleus (PPN) neurons. J Neural Transm (Vienna) 2013; 120:1027-38. [PMID: 23263542 PMCID: PMC3618992 DOI: 10.1007/s00702-012-0957-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/10/2012] [Indexed: 12/24/2022]
Abstract
Leptin, a hormone that regulates appetite and energy expenditure, is increased in obese individuals, although these individuals often exhibit leptin resistance. Obesity is characterized by sleep/wake disturbances, such as excessive daytime sleepiness, increased REM sleep, increased nighttime arousals, and decreased percentage of total sleep time. Several studies have shown that short sleep duration is highly correlated with decreased leptin levels in both animal and human models. Arousal and rapid eye movement (REM) sleep are regulated by the cholinergic arm of the reticular activating system, the pedunculopontine nucleus (PPN). The goal of this project was to determine the role of leptin in the PPN, and thus in obesity-related sleep disorders. Whole-cell patch-clamp recordings were conducted on PPN neurons in 9- to 17-day-old rat brainstem slices. Leptin decreased action potential (AP) amplitude, AP frequency, and h-current (I(H)). These findings suggest that leptin causes a blockade of Na⁺ channels. Therefore, we conducted an experiment to test the effects of leptin on Na⁺ conductance. To determine the average voltage dependence of this conductance, results from each cell were equally weighted by expressing conductance as a fraction of the maximum conductance in each cell. I Na amplitude was decreased in a dose-dependent manner, suggesting a direct effect of leptin on these channels. The average decrease in Na⁺ conductance by leptin was ~40 %. We hypothesize that leptin normally decreases activity in the PPN by reducing I(H) and I(Na) currents, and that in states of leptin dysregulation (i.e., leptin resistance) this effect may be blunted, therefore causing increased arousal and REM sleep drive, and ultimately leading to sleep-related disorders.
Collapse
Affiliation(s)
- Paige Beck
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - D. Keith Williams
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Edgar Garcia-Rill
- Center for Translational Neuroscience, Dept. Neurobiology & Dev. Sci., University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
24
|
Nieh EH, Kim SY, Namburi P, Tye KM. Optogenetic dissection of neural circuits underlying emotional valence and motivated behaviors. Brain Res 2013; 1511:73-92. [PMID: 23142759 PMCID: PMC4099056 DOI: 10.1016/j.brainres.2012.11.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 11/01/2012] [Accepted: 11/02/2012] [Indexed: 12/26/2022]
Abstract
The neural circuits underlying emotional valence and motivated behaviors are several synapses away from both defined sensory inputs and quantifiable motor outputs. Electrophysiology has provided us with a suitable means for observing neural activity during behavior, but methods for controlling activity for the purpose of studying motivated behaviors have been inadequate: electrical stimulation lacks cellular specificity and pharmacological manipulation lacks temporal resolution. The recent emergence of optogenetic tools provides a new means for establishing causal relationships between neural activity and behavior. Optogenetics, the use of genetically-encodable light-activated proteins, permits the modulation of specific neural circuit elements with millisecond precision. The ability to control individual cell types, and even projections between distal regions, allows us to investigate functional connectivity in a causal manner. The greatest consequence of controlling neural activity with finer precision has been the characterization of individual neural circuits within anatomical brain regions as defined functional units. Within the mesolimbic dopamine system, optogenetics has helped separate subsets of dopamine neurons with distinct functions for reward, aversion and salience processing, elucidated GABA neuronal effects on behavior, and characterized connectivity with forebrain and cortical structures. Within the striatum, optogenetics has confirmed the opposing relationship between direct and indirect pathway medium spiny neurons (MSNs), in addition to characterizing the inhibition of MSNs by cholinergic interneurons. Within the hypothalamus, optogenetics has helped overcome the heterogeneity in neuronal cell-type and revealed distinct circuits mediating aggression and feeding. Within the amygdala, optogenetics has allowed the study of intra-amygdala microcircuitry as well as interconnections with distal regions involved in fear and anxiety. In this review, we will present the body of optogenetic studies that has significantly enhanced our understanding of emotional valence and motivated behaviors. This article is part of a Special Issue entitled Optogenetics (7th BRES).
Collapse
Affiliation(s)
- Edward H. Nieh
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sung-Yon Kim
- Department of Bioengineering, Neurosciences Program, Stanford University, Stanford, CA, USA
| | - Praneeth Namburi
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kay M. Tye
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
25
|
Thanos PK, Robison L, Nestler EJ, Kim R, Michaelides M, Lobo MK, Volkow ND. Mapping brain metabolic connectivity in awake rats with μPET and optogenetic stimulation. J Neurosci 2013; 33:6343-9. [PMID: 23575833 PMCID: PMC3666931 DOI: 10.1523/jneurosci.4997-12.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 12/23/2012] [Accepted: 02/18/2013] [Indexed: 12/28/2022] Open
Abstract
Positron emission tomography (PET) with [(18)F]2-fluoro-2-deoxy-D-glucose was used to measure changes in regional brain glucose metabolism (BGluM) in response to optogenetic stimulation (using the excitatory channelrhodopsin-2) of the nucleus accumbens (NAc) in awake rats. We demonstrated not only increases in BGluM that correlated with c-Fos expression in the region of stimulation, but also BGluM increases in the ipsilateral striatum, periaqueductal gray, and somatosensory cortex, and in contralateral amygdala, ventral pallidum, globus pallidus, and hippocampus, as well as decreases in BGluM in regions of the default mode network (retrosplenial cortex and cingulate gyrus) and secondary motor cortex. Additional exploration of c-Fos expression in regions found to be activated by PET results found corroborating evidence, with increased c-Fos expression in the ipsilateral somatosensory cortex, contralateral amygdala and globus pallidus, and bilateral periaqueductal gray. These findings are consistent with optogenetic excitation of the area of stimulation (NAc), as well as with stimulatory and inhibitory effects on downstream regions. They also confirm the utility of PET imaging to monitor connectivity in the awake rodent brain.
Collapse
Affiliation(s)
- Panayotis K. Thanos
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Medical Department, Brookhaven National Laboratory, Upton, New York 11973
- Department of Psychology, Stony Brook University, Stony Brook New York 11794
| | - Lisa Robison
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852
- Department of Psychology, Stony Brook University, Stony Brook New York 11794
| | - Eric J. Nestler
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Ronald Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Michael Michaelides
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029
| | - Mary-Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, and
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852
| |
Collapse
|
26
|
Abstract
Over more than a century of research has established the fact that sleep benefits the retention of memory. In this review we aim to comprehensively cover the field of "sleep and memory" research by providing a historical perspective on concepts and a discussion of more recent key findings. Whereas initial theories posed a passive role for sleep enhancing memories by protecting them from interfering stimuli, current theories highlight an active role for sleep in which memories undergo a process of system consolidation during sleep. Whereas older research concentrated on the role of rapid-eye-movement (REM) sleep, recent work has revealed the importance of slow-wave sleep (SWS) for memory consolidation and also enlightened some of the underlying electrophysiological, neurochemical, and genetic mechanisms, as well as developmental aspects in these processes. Specifically, newer findings characterize sleep as a brain state optimizing memory consolidation, in opposition to the waking brain being optimized for encoding of memories. Consolidation originates from reactivation of recently encoded neuronal memory representations, which occur during SWS and transform respective representations for integration into long-term memory. Ensuing REM sleep may stabilize transformed memories. While elaborated with respect to hippocampus-dependent memories, the concept of an active redistribution of memory representations from networks serving as temporary store into long-term stores might hold also for non-hippocampus-dependent memory, and even for nonneuronal, i.e., immunological memories, giving rise to the idea that the offline consolidation of memory during sleep represents a principle of long-term memory formation established in quite different physiological systems.
Collapse
Affiliation(s)
- Björn Rasch
- Division of Biopsychology, Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
27
|
Abstract
The circadian pattern of seizures in people with epilepsy (PWE) was first described two millennia ago. However, these phenomena have not received enough scientific attention, possibly due to the lack of promising hypotheses to address the interaction between seizure generation and a physiological clock. To propose testable hypotheses at the molecular level, interactions between circadian rhythm, especially transcription factors governing clock genes expression, and the mTOR (mammalian target of rapamycin) signaling pathway, the major signaling pathway in epilepsy, will be reviewed. Then, two closely related hypotheses will be proposed: (1) Rhythmic activity of hyperactivated mTOR signaling molecules results in rhythmic increases in neuronal excitability. These rhythmic increases in excitability periodically exceed the seizure threshold, displaying the behavioral seizures. (2) Oscillation of neuronal excitability in SCN modulates the rhythmic excitability in the hippocampus through subiculum via long-range projections. Findings from published results, their implications, and proposals for new experiments will be discussed. These attempts may ignite further discussion on what we still need to learn about the rhythmicity of spontaneous seizures.
Collapse
Affiliation(s)
- Chang-Hoon Cho
- Epilepsy Research Laboratory, Department of Pediatrics, Children's Hospital of Philadelphia Philadelphia, PA, USA
| |
Collapse
|
28
|
|
29
|
Zhao P, Shao Y, Zhang M, Fan K, Kong X, Wang R, Hou Y. Neuropeptide S promotes wakefulness through activation of the posterior hypothalamic histaminergic and orexinergic neurons. Neuroscience 2012; 207:218-26. [DOI: 10.1016/j.neuroscience.2012.01.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 11/29/2011] [Accepted: 01/09/2012] [Indexed: 10/14/2022]
|
30
|
Urbano FJ, Kezunovic N, Hyde J, Simon C, Beck P, Garcia-Rill E. Gamma band activity in the reticular activating system. Front Neurol 2012; 3:6. [PMID: 22319508 PMCID: PMC3269033 DOI: 10.3389/fneur.2012.00006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/06/2012] [Indexed: 12/24/2022] Open
Abstract
This review considers recent evidence showing that cells in three regions of the reticular activating system (RAS) exhibit gamma band activity, and describes the mechanisms behind such manifestation. Specifically, we discuss how cells in the mesopontine pedunculopontine nucleus (PPN), intralaminar parafascicular nucleus (Pf), and pontine subcoeruleus nucleus dorsalis (SubCD) all fire in the beta/gamma band range when maximally activated, but no higher. The mechanisms behind this ceiling effect have been recently elucidated. We describe recent findings showing that every cell in the PPN have high-threshold, voltage-dependent P/Q-type calcium channels that are essential, while N-type calcium channels are permissive, to gamma band activity. Every cell in the Pf also showed that P/Q-type and N-type calcium channels are responsible for this activity. On the other hand, every SubCD cell exhibited sodium-dependent subthreshold oscillations. A novel mechanism for sleep–wake control based on well-known transmitter interactions, electrical coupling, and gamma band activity is described. The data presented here on inherent gamma band activity demonstrates the global nature of sleep–wake oscillation that is orchestrated by brainstem–thalamic mechanism, and questions the undue importance given to the hypothalamus for regulation of sleep–wakefulness. The discovery of gamma band activity in the RAS follows recent reports of such activity in other subcortical regions like the hippocampus and cerebellum. We hypothesize that, rather than participating in the temporal binding of sensory events as seen in the cortex, gamma band activity manifested in the RAS may help stabilize coherence related to arousal, providing a stable activation state during waking and paradoxical sleep. Most of our thoughts and actions are driven by pre-conscious processes. We speculate that continuous sensory input will induce gamma band activity in the RAS that could participate in the processes of pre-conscious awareness, and provide the essential stream of information for the formulation of many of our actions.
Collapse
Affiliation(s)
- Francisco J Urbano
- Instituto de Fisiología, Biología Molecular y Neurociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, University of Buenos Aires Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
31
|
Banghart MR, Sabatini BL. Photoactivatable neuropeptides for spatiotemporally precise delivery of opioids in neural tissue. Neuron 2012; 73:249-59. [PMID: 22284180 PMCID: PMC3282187 DOI: 10.1016/j.neuron.2011.11.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2011] [Indexed: 12/19/2022]
Abstract
Neuropeptides activate G protein-coupled receptors to acutely modulate cellular excitability and synaptic transmission. However, due to the lack of reagents for precise delivery of peptides within dense brain tissue, the spatiotemporal scale over which neuropeptides act is unknown. To achieve rapid and spatially delimited delivery of neuropeptides in mammalian brain tissue, we developed photoactivatable analogs of two opioids: [Leu⁵]-enkephalin (LE) and the 8 amino acid form of Dynorphin A (Dyn-8). These peptides are functionally inactive prior to photolysis, and exposure to ultraviolet (UV) light causes clean release of LE and Dyn-8. Recordings from acute slices of rat locus coeruleus (LC) demonstrated that photorelease of LE activates mu opioid receptor-coupled K+ channels with kinetics that approach the limits imposed by G protein-mediated signaling. Temporally precise and spatially delimited photorelease revealed the kinetics and ionic nature of the mu opioid response and the mechanisms that determine the spatial profile of enkephalinergic volume transmission in LC.
Collapse
Affiliation(s)
- Matthew R. Banghart
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bernardo L. Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
32
|
Meier JC, Harvey RJ, Seeburg P. Frontiers in molecular neuroscience - résumé and perspective. Front Mol Neurosci 2012; 4:58. [PMID: 22232574 PMCID: PMC3248788 DOI: 10.3389/fnmol.2011.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 12/15/2011] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jochen C Meier
- Max Delbrück Center for Molecular Medicine Berlin, Germany
| | | | | |
Collapse
|
33
|
Abstract
Optogenetics is a rapidly evolving field of technology that allows optical control of genetically targeted biological systems at high temporal and spatial resolution. By heterologous expression of light-sensitive microbial membrane proteins, opsins, cell type-specific depolarization or silencing can be optically induced on a millisecond time scale. What started in a petri dish is applicable today to more complex systems, ranging from the dissection of brain circuitries in vitro to behavioral analyses in freely moving animals. Persistent technical improvement has focused on the identification of new opsins, suitable for optogenetic purposes and genetic engineering of existing ones. Optical stimulation can be combined with various readouts defined by the desired resolution of the experimental setup. Although recent developments in optogenetics have largely focused on neuroscience it has lately been extended to other targets, including stem cell research and regenerative medicine. Further development of optogenetic approaches will not only highly increase our insight into health and disease states but might also pave the way for a future use in therapeutic applications.
Collapse
|
34
|
Expanding neurotransmitters in the hypothalamic neurocircuitry for energy balance regulation. Protein Cell 2011; 2:800-13. [PMID: 22058035 DOI: 10.1007/s13238-011-1112-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 10/10/2011] [Indexed: 01/06/2023] Open
Abstract
The current epidemic of obesity and its associated metabolic syndromes impose unprecedented challenges to our society. Despite intensive research on obesity pathogenesis, an effective therapeutic strategy to treat and cure obesity is still lacking. Exciting studies in last decades have established the importance of the leptin neural pathway in the hypothalamus in the regulation of body weight homeostasis. Important hypothalamic neuropeptides have been identified as critical neurotransmitters from leptin-sensitive neurons to mediate leptin action. Recent research advance has significantly expanded the list of neurotransmitters involved in body weight-regulating neural pathways, including fast-acting neurotransmitters, gamma-aminobutyric acid (GABA) and glutamate. Given the limited knowledge on the leptin neural pathway for body weight homeostasis, understanding the function of neurotransmitters released from key neurons for energy balance regulation is essential for delineating leptin neural pathway and eventually for designing effective therapeutic drugs against the obesity epidemic.
Collapse
|
35
|
Robertson HR, Feng G. Annual Research Review: Transgenic mouse models of childhood-onset psychiatric disorders. J Child Psychol Psychiatry 2011; 52:442-75. [PMID: 21309772 PMCID: PMC3075087 DOI: 10.1111/j.1469-7610.2011.02380.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Childhood-onset psychiatric disorders, such as attention deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), mood disorders, obsessive compulsive spectrum disorders (OCSD), and schizophrenia (SZ), affect many school-age children, leading to a lower quality of life, including difficulties in school and personal relationships that persist into adulthood. Currently, the causes of these psychiatric disorders are poorly understood, resulting in difficulty diagnosing affected children, and insufficient treatment options. Family and twin studies implicate a genetic contribution for ADHD, ASD, mood disorders, OCSD, and SZ. Identification of candidate genes and chromosomal regions associated with a particular disorder provide targets for directed research, and understanding how these genes influence the disease state will provide valuable insights for improving the diagnosis and treatment of children with psychiatric disorders. Transgenic mouse models are one important approach in the study of human diseases, allowing for the use of a variety of experimental approaches to dissect the contribution of a specific chromosomal or genetic abnormality in human disorders. While it is impossible to model an entire psychiatric disorder in a single mouse model, these models can be extremely valuable in dissecting out the specific role of a gene, pathway, neuron subtype, or brain region in a particular abnormal behavior. In this review we discuss existing transgenic mouse models for childhood-onset psychiatric disorders. We compare the strength and weakness of various transgenic mouse models proposed for each of the common childhood-onset psychiatric disorders, and discuss future directions for the study of these disorders using cutting-edge genetic tools.
Collapse
Affiliation(s)
- Holly R. Robertson
- Duke University, Neurobiology Department Durham, N.C.,Massachusetts Institute of Technology, Brain and Cognitive Sciences Department Cambridge, M.A
| | - Guoping Feng
- Duke University, Neurobiology Department Durham, N.C.,Massachusetts Institute of Technology, Brain and Cognitive Sciences Department Cambridge, M.A
| |
Collapse
|
36
|
Fenzl T, Romanowski CPN, Flachskamm C, Deussing JM, Kimura M. Wake-promoting effects of orexin: Its independent actions against the background of an impaired corticotropine-releasing hormone receptor system. Behav Brain Res 2011; 222:43-50. [PMID: 21420442 DOI: 10.1016/j.bbr.2011.03.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 03/04/2011] [Accepted: 03/11/2011] [Indexed: 01/08/2023]
Abstract
It is widely accepted that orexin (hypocretin) bears wake-promoting effects. While under normal conditions the circadian rhythm of orexin release has a clear circadian distribution, the amplitude of orexin fluctuation is dampened in depression. Interestingly, clinical symptoms of depression include several sleep disturbances. In this disease, corticotropin-releasing hormone (CRH) seems to be another factor influencing sleep. As neurophysiological interactions and anatomical connections between the orexinergic and the CRH system point to mutual influences of these two neuropeptides, we examined whether a dysfunctional CRH-receptor system in two different CRH receptor knock out models alters general wake-promoting effects of orexin applied exogenously. Orexin was injected intracerebroventricularlly into CNS-restricted CRH-receptor type 1 knockout mice (CRH-R1 KO) and CRH-receptor type 2 knockout mice (CRH-R2 KO) and baseline sleep was recorded from the freely behaving mice. A third experiment included antisauvagine-30 injections (CRH-R2 antagonist) into CRH-R1 KO animals. Orexin had similar wake-promoting effects in CRH-R1KO mice, in CRH-R2 KO animals and in CRH-R1KO mice treated with antisauvagine-30. Consistent results were obtained from all corresponding control littermate experiments. According to our results we conclude that the wake-promoting effects of orexin are not influenced by a possible contribution of CRH.
Collapse
Affiliation(s)
- Thomas Fenzl
- Max-Planck-Institute of Psychiatry, Kraepelinstrasse 2, 80804 Munich, Germany.
| | | | | | | | | |
Collapse
|
37
|
DEURVEILHER S, SEMBA K. Basal forebrain regulation of cortical activity and sleep-wake states: Roles of cholinergic and non-cholinergic neurons. Sleep Biol Rhythms 2011. [DOI: 10.1111/j.1479-8425.2010.00465.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Affiliation(s)
- William Wisden
- Division of Cell and Molecular Biology, Imperial College London London, UK
| | | |
Collapse
|