Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Sleep ; 47(5)2024 May 10.
Article in English | MEDLINE | ID: mdl-38447008

ABSTRACT

Dynorphin is an endogenous opiate localized in many brain regions and spinal cord, but the activity of dynorphin neurons during sleep is unknown. Dynorphin is an inhibitory neuropeptide that is coreleased with orexin, an excitatory neuropeptide. We used microendoscopy to test the hypothesis that, like orexin, the dynorphin neurons are wake-active. Dynorphin-cre mice (n = 3) were administered rAAV8-Ef1a-Con/Foff 2.0-GCaMP6M into the zona incerta-perifornical area, implanted with a GRIN lens (gradient reflective index), and electrodes to the skull that recorded sleep. One month later, a miniscope imaged calcium fluorescence in dynorphin neurons during multiple bouts of wake, non-rapid-eye movement (NREM), and rapid-eye movement (REM) sleep. Unbiased data analysis identified changes in calcium fluorescence in 64 dynorphin neurons. Most of the dynorphin neurons (72%) had the highest fluorescence during bouts of active and quiet waking compared to NREM or REM sleep; a subset (20%) were REM-max. Our results are consistent with the emerging evidence that the activity of orexin neurons can be classified as wake-max or REM-max. Since the two neuropeptides are coexpressed and coreleased, we suggest that dynorphin-cre-driven calcium sensors could increase understanding of the role of this endogenous opiate in pain and sleep.


Subject(s)
Dynorphins , Neurons , Sleep, REM , Wakefulness , Zona Incerta , Animals , Mice , Dynorphins/metabolism , Dynorphins/physiology , Neurons/physiology , Zona Incerta/physiology , Zona Incerta/physiopathology , Sleep, REM/physiology , Wakefulness/physiology , Male , Orexins/metabolism , Orexins/physiology
2.
Mol Psychiatry ; 29(2): 327-341, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38123729

ABSTRACT

Hypocretin/Orexin (HCRT/OX) and dopamine (DA) are both key effectors of salience processing, reward and stress-related behaviors and motivational states, yet their respective roles and interactions are poorly delineated. We inactivated HCRT-to-DA connectivity by genetic disruption of Hypocretin receptor-1 (Hcrtr1), Hypocretin receptor-2 (Hcrtr2), or both receptors (Hcrtr1&2) in DA neurons and analyzed the consequences on vigilance states, brain oscillations and cognitive performance in freely behaving mice. Unexpectedly, loss of Hcrtr2, but not Hcrtr1 or Hcrtr1&2, induced a dramatic increase in theta (7-11 Hz) electroencephalographic (EEG) activity in both wakefulness and rapid-eye-movement sleep (REMS). DAHcrtr2-deficient mice spent more time in an active (or theta activity-enriched) substate of wakefulness, and exhibited prolonged REMS. Additionally, both wake and REMS displayed enhanced theta-gamma phase-amplitude coupling. The baseline waking EEG of DAHcrtr2-deficient mice exhibited diminished infra-theta, but increased theta power, two hallmarks of EEG hyperarousal, that were however uncoupled from locomotor activity. Upon exposure to novel, either rewarding or stress-inducing environments, DAHcrtr2-deficient mice featured more pronounced waking theta and fast-gamma (52-80 Hz) EEG activity surges compared to littermate controls, further suggesting increased alertness. Cognitive performance was evaluated in an operant conditioning paradigm, which revealed that DAHcrtr2-ablated mice manifest faster task acquisition and higher choice accuracy under increasingly demanding task contingencies. However, the mice concurrently displayed maladaptive patterns of reward-seeking, with behavioral indices of enhanced impulsivity and compulsivity. None of the EEG changes observed in DAHcrtr2-deficient mice were seen in DAHcrtr1-ablated mice, which tended to show opposite EEG phenotypes. Our findings establish a clear genetically-defined link between monosynaptic HCRT-to-DA neurotransmission and theta oscillations, with a differential and novel role of HCRTR2 in theta-gamma cross-frequency coupling, attentional processes, and executive functions, relevant to disorders including narcolepsy, attention-deficit/hyperactivity disorder, and Parkinson's disease.


Subject(s)
Cognition , Dopaminergic Neurons , Electroencephalography , Orexin Receptors , Wakefulness , Animals , Mice , Dopaminergic Neurons/physiology , Dopaminergic Neurons/metabolism , Cognition/physiology , Orexin Receptors/metabolism , Orexin Receptors/physiology , Wakefulness/physiology , Male , Electroencephalography/methods , Arousal/physiology , Mice, Inbred C57BL , Mice, Knockout , Orexins/metabolism , Orexins/physiology , Sleep, REM/physiology , Signal Transduction/physiology , Theta Rhythm/physiology , Reward , Dopamine/metabolism
3.
Proc Natl Acad Sci U S A ; 119(16): e2113518119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412900

ABSTRACT

Fear is essential for survival, but excessive anxiety behavior is debilitating. Anxiety disorders affecting millions of people are a global health problem, where new therapies and targets are much needed. Deep brain stimulation (DBS) is established as a therapy in several neurological disorders, but is underexplored in anxiety disorders. The lateral hypothalamus (LH) has been recently revealed as an origin of anxiogenic brain signals, suggesting a target for anxiety treatment. Here, we develop and validate a DBS strategy for modulating anxiety-like symptoms by targeting the LH. We identify a DBS waveform that rapidly inhibits anxiety-implicated LH neural activity and suppresses innate and learned anxiety behaviors in a variety of mouse models. Importantly, we show that the LH DBS displays high temporal and behavioral selectivity: Its affective impact is fast and reversible, with no evidence of side effects such as impaired movement, memory loss, or epileptic seizures. These data suggest that acute hypothalamic DBS could be a useful strategy for managing treatment-resistant anxiety disorders.


Subject(s)
Anxiety Disorders , Deep Brain Stimulation , Hypothalamic Area, Lateral , Animals , Anxiety Disorders/therapy , Deep Brain Stimulation/methods , Mice , Orexins/antagonists & inhibitors , Orexins/physiology
4.
Science ; 375(6583): eabh3021, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35201886

ABSTRACT

Sleep quality declines with age; however, the underlying mechanisms remain elusive. We found that hyperexcitable hypocretin/orexin (Hcrt/OX) neurons drive sleep fragmentation during aging. In aged mice, Hcrt neurons exhibited more frequent neuronal activity epochs driving wake bouts, and optogenetic activation of Hcrt neurons elicited more prolonged wakefulness. Aged Hcrt neurons showed hyperexcitability with lower KCNQ2 expression and impaired M-current, mediated by KCNQ2/3 channels. Single-nucleus RNA-sequencing revealed adaptive changes to Hcrt neuron loss in the aging brain. Disruption of Kcnq2/3 genes in Hcrt neurons of young mice destabilized sleep, mimicking aging-associated sleep fragmentation, whereas the KCNQ-selective activator flupirtine hyperpolarized Hcrt neurons and rejuvenated sleep architecture in aged mice. Our findings demonstrate a mechanism underlying sleep instability during aging and a strategy to improve sleep continuity.


Subject(s)
Aging , Neurons/physiology , Orexins/physiology , Sleep Deprivation/physiopathology , Sleep , Wakefulness , Aminopyridines/pharmacology , Animals , CRISPR-Cas Systems , Electroencephalography , Electromyography , Female , Hypothalamic Area, Lateral/physiopathology , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism , Male , Mice , Narcolepsy/genetics , Narcolepsy/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways , Optogenetics , Patch-Clamp Techniques , RNA-Seq , Sleep Quality
5.
Neurotox Res ; 39(6): 2141-2153, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34495449

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease affecting about 2% of the population. A neuropeptide, orexin, is linked with sleep abnormalities in the parkinsonian patient. This study aimed to review the changes in the orexinergic system in parkinsonian subjects and the effects of orexin. A number of search techniques were used and presumed during the search, including cloud databank searches of PubMed and Medline using title words, keywords, and MeSH terms. PD is characterised by motor dysfunctions (postural instability, rigidity, tremor) and cognitive disorders, sleep-wake abnormalities grouped under non-motor disorders. The Orexinergic system found in the hypothalamus is linked with autonomic function, neuroprotection, learning and memory, and the sleep-wake cycle. Prepro-orexin, a precursor peptide (130 amino acids), gives rise to orexins (Orx-A and Orx-B). Serum orexin level measurement is vital for evaluating several neurological disorders (Alzheimer's disease, Huntington's disease, and PD). Orexinergic neurons are activated by hypoglycemia and ghrelin, while they are restrained by food consumption and leptin. Orexinergic system dysfunctioning was found to be linked with non-motor symptoms (sleep abnormalities) in PD. Orexinergic neuron's behaviour may be either inhibitory or excitatory depending on the environment in which they are present. As well, orexin antagonists are found to improve the abnormal sleep pattern. Since the orexinergic system plays a role in several psychological and neurological disorders, therefore, these disorders can be managed by targeting this system.


Subject(s)
Neurons/metabolism , Orexins/physiology , Parkinson Disease/etiology , Animals , Humans , Neurons/pathology , Orexins/metabolism , Parkinson Disease/pathology
6.
Molecules ; 26(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34577105

ABSTRACT

Granulosa cells (GCs) are essential for follicular growth, development, and atresia. The orexin-A (OXA) neuropeptide is widely involved in the regulation of various biological functions. OXA selectively binds to orexin receptor type 1 (OX1R) and mediates all its biological actions via OX1R. This study aimed to explore the expression of OXA and OX1R and their regulatory role in GCs proliferation, cell cycle progression, apoptosis, oocyte maturation, and underlying molecular mechanisms of these processes and elucidate its novel signaling pathway. Western blotting and RT-qPCR showed that OXA and OX1R were expressed during different developmental stages of GCs, and siRNA transfection successfully inhibited the expression of OX1R at the translational and transcriptional levels. Flow cytometry revealed that OX1R knockdown upregulated GCs apoptosis and triggered S-phase arrest in cell cycle progression. RT-qPCR and Western blotting showed significantly reduced expression of Bcl-2 and elevated expression of Bax, caspase-3, TNF-α, and P21 in OX1R-silenced GCs. Furthermore, the CCK-8 assay showed that knockdown of OX1R suppressed GCs proliferation by downregulating the expression of PCNA, a proliferation marker gene, at the translational and transcriptional levels. Western blotting revealed that knockdown of OX1R resulted in a considerable decrease of the phosphorylation level of the AKT and ERK1/2 proteins, indicating that the AKT/ERK1/2 pathway is involved in regulating GCs proliferation and apoptosis. In addition, OX1R silencing enhanced the mRNA expression of GDF9 and suppressed the mRNA expression of BMP15 in mouse GCs. Collectively, these results reveal a novel regulatory role of OXA in the development of GCs and folliculogenesis by regulating proliferation, apoptosis, and cell cycle progression. Therefore, OXA can be a promising therapeutic agent for female infertility.


Subject(s)
Granulosa Cells/metabolism , MAP Kinase Signaling System/drug effects , Orexins/physiology , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Down-Regulation/genetics , Female , Granulosa Cells/drug effects , Granulosa Cells/physiology , MAP Kinase Signaling System/genetics , Mice , Orexin Receptors/genetics , Orexin Receptors/metabolism , Orexins/metabolism , Ovarian Follicle/drug effects , Primary Cell Culture
7.
Peptides ; 145: 170629, 2021 11.
Article in English | MEDLINE | ID: mdl-34416308

ABSTRACT

Hypothalamic neurons that produce the peptide transmitters orexins/hypocretins (HONs) broadcast their predominantly neuroexcitatory outputs to the entire brain via their extremely wide axonal projections. HONs were originally reported to be activated by food deprivation, and to stimulate arousal, energy expenditure, and eating. This led to extensive studies of HONs in the context of nutrient-sensing and energy balance control. While activation of HONs by body energy depletion continues to be supported by experimental evidence, it has also become clear that HONs are robustly activated not only by nutrient depletion, but also by diverse sensory stimuli (both neutral and those associated with rewarding or aversive events), seemingly unrelated to each other or to energy balance. One theory that could unify these findings is that all these stimuli signal "stress" - defined either as a potentially harmful state, or an awareness of reward deficiency. If HON activity is conceptualized as a cumulative representation of stress, then many of the reported HONs outputs - including EEG arousal, sympathetic activation, place avoidance, and exploratory behaviours - could be viewed as logical stress-counteracting responses. We discuss evidence for and against this unifying theory of HON function, including the alterations in HON activity observed in anxiety and depression disorders. We propose that, in order to orchestrate stress-countering responses, HONs need to coactivate motivation and aversion brain systems, and the impact of HON stimulation on affective states may be perceived as rewarding or aversive depending on the baseline HON activity.


Subject(s)
Brain/physiology , Neurons/physiology , Orexins/metabolism , Stress, Physiological/physiology , Animals , Anxiety/metabolism , Depression/metabolism , Energy Metabolism , Exploratory Behavior , Humans , Motivation , Orexins/physiology , Reward
8.
Metab Brain Dis ; 36(7): 1419-1444, 2021 10.
Article in English | MEDLINE | ID: mdl-34224065

ABSTRACT

Orexin (hypocretin), is a neuropeptide produced by a subset of neurons in the lateral hypothalamus. From the lateral hypothalamus, the orexin-containing neurons project their fibres extensively to other brain structures, and the spinal cord constituting the central orexinergic system. Generally, the term ''orexinergic system'' usually refers to the orexin peptides and their receptors, as well as to the orexin neurons and their projections to different parts of the central nervous system. The extensive networks of orexin axonal fibres and their terminals allow these neuropeptidergic neurons to exert great influence on their target regions. The hypothalamic neurons containing the orexin neuropeptides have been implicated in diverse functions, especially related to the control of a variety of homeostatic functions including feeding behaviour, arousal, wakefulness stability and energy expenditure. The broad range of functions regulated by the orexinergic system has led to its description as ''physiological integrator''. In the last two decades, the orexinergic system has been a topic of great interest to the scientific community with many reports in the public domain. From the documentations, variations exist in the neuroanatomical profile of the orexinergic neuron soma, fibres and their receptors from animal to animal. Hence, this review highlights the distinct variabilities in the morphophysiological aspects of the orexinergic system in the vertebrate animals, mammals and non-mammals, its presence in other brain-related structures, including its involvement in ageing and neurodegenerative diseases. The presence of the neuropeptide in the cerebrospinal fluid and peripheral tissues, as well as its alteration in different animal models and conditions are also reviewed.


Subject(s)
Brain/physiology , Neurons/physiology , Orexin Receptors/physiology , Orexins/physiology , Aging/physiology , Animals , Humans , Orexins/cerebrospinal fluid , Signal Transduction/physiology , Spinal Cord/physiology
9.
J Pharmacol Sci ; 146(4): 183-191, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34116731

ABSTRACT

Short chain fatty acids readily crosses the gut-blood and blood-brain barrier and acts centrally to influence neuronal signaling. We hypothesized that butyrate, a short-chain fatty acid produced by bacterial fermentation, in the central nervous system may play a role in the regulation of intestinal functions. Colonic permeability and visceral sensation was evaluated in rats. Septic lethality was evaluated in a sepsis model induced by subcutaneous administration of both lipopolysaccharide and colchicine. Intracisternal butyrate dose-dependently improved colonic hyperpermeability and visceral nociception. In contrast, subcutaneous injection of butyrate failed to change it. Intracisternal orexin 1 receptor antagonist or surgical vagotomy blocked the central butyrate-induced improvement of colonic hyperpermeability. The improvement of intestinal hyperpermeability by central butyrate or intracisternal orexin-A was blocked by cannabinoid 1 or 2 receptor antagonist. Intracisternal butyrate significantly improved survival period in septic rats. These results suggest that butyrate acts in the central nervous system to improve gut permeability and visceral nociception through cannabinoid signaling. Endogenous orexin in the brain may mediate the reduction of intestinal hyperpermeability by central butyrate through the vagus nerve. We would suggest that improvement of leaky gut by central butyrate may induce visceral antinociception and protection from septic lethality.


Subject(s)
Butyrates/pharmacology , Colon/metabolism , Fatty Acids, Volatile/pharmacology , Nociception/drug effects , Shock, Septic/mortality , Viscera/physiology , Animals , Brain/metabolism , Butyrates/administration & dosage , Colchicine/adverse effects , Disease Models, Animal , Fatty Acids, Volatile/administration & dosage , Lipopolysaccharides/adverse effects , Male , Orexins/metabolism , Orexins/physiology , Permeability , Rats, Sprague-Dawley , Shock, Septic/chemically induced , Shock, Septic/prevention & control , Viscera/drug effects
10.
Front Neurol Neurosci ; 45: 128-138, 2021.
Article in English | MEDLINE | ID: mdl-34052807

ABSTRACT

Sleep is one of the pillars of health. Experimental models of acute sleep loss, of chronic partial sleep deprivation, and of sleep fragmentation in healthy sleepers are helpful models of sleep deficiency produced by insufficient sleep duration, sleep timing, and sleep disorders. Sleep deficiency is associated with changes in markers associated with risk for disease. These include metabolic, inflammatory, and autonomic markers of risk. In addition, sleep disruption and sleep deficits lead to mood instability, lack of positive outlook, and impaired neurobehavioral functioning. On a population level, insufficient sleep is associated with increased risk for hypertension and diabetes. Sleep disturbance is very common, and about half the population will report that they have experienced insomnia at some time in their lives. Approximately 10% of the population describe daytime impairment due to sleep disturbance at night, consistent with a diagnosis of insomnia disorder. The hypothalamic neuropeptides, orexin-A and orexin-B, act through G-protein-coupled receptors (orexin-1 and orexin-2 receptors). Dual and selective orexin-2 receptor antagonists have shown efficacy in inducing sleep in men and women with insomnia disorder by accelerating sleep onset and improving sleep efficiency and total sleep time. Further study comparing these medications, in short- and longer-term use models, is recommended. Greater understanding of comparative effects on mood, neurobehavioral, and physiological systems will help determine the extent of clinical utility of dual versus selective orexin receptor antagonists.


Subject(s)
Cardiovascular Diseases , Chronic Pain , Metabolic Diseases , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/physiology , Orexins/physiology , Sleep Deprivation , Sleep Initiation and Maintenance Disorders , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Chronic Pain/etiology , Chronic Pain/metabolism , Chronic Pain/physiopathology , Humans , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Sleep Deprivation/complications , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep Initiation and Maintenance Disorders/complications , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep Initiation and Maintenance Disorders/metabolism , Sleep Initiation and Maintenance Disorders/physiopathology
11.
Front Neurol Neurosci ; 45: 52-60, 2021.
Article in English | MEDLINE | ID: mdl-34052808

ABSTRACT

Hypothalamic hypocretin/orexin neurons have been initially conceptualized as slow, modulatory controllers of behavior. Furthermore, their behavioral effects have been assumed to be a secondary consequence of their impact on arousal. However, cellular-resolution calcium imaging and optogenetic studies show that orexin neurons regulate self-generated and sensory-evoked movement on rapid, subsecond timescales. Orexin cell activity rapidly and transiently peaks before and during movements. Optogenetic prevention of this activation reduces the probability of locomotion initiation, and optogenetic mimicry of orexin cell activation rapidly causes locomotion. Neural ensemble calcium imaging experiments reveal that the same orexin cells whose activity underlies movement initiation display subsecond-latency responses to diverse sensory stimuli. These findings establish orexin neurons as rapid and strong sensorimotor controllers that are in many ways operationally similar to classic subcortical movement controllers, such as midbrain dopamine neurons. While a scientific definition of "arousal" is still lacking, the subsecond-scale sensorimotor control by orexin neurons could be viewed as reminiscent of a motor rather than an arousal system.


Subject(s)
Hypothalamus/physiology , Locomotion/physiology , Motor Activity/physiology , Neurons/physiology , Orexins/physiology , Sensation/physiology , Animals , Humans
12.
Front Neurol Neurosci ; 45: 1-10, 2021.
Article in English | MEDLINE | ID: mdl-34052811

ABSTRACT

The discovery of the hypocretins/orexins (HCRTs) has revolutionized sleep science in the last two decades. A combination of anatomical tracing methods, optogenetics, and pharmacology is delineating a blueprint of functional inputs and outputs of the HCRT system. Here, we discuss several models of HCRT action that involve the integration between physiological variables, circadian output, and sleep homeostasis. Generation of activity maps during the sleep-wake cycle at the cellular level will allow investigators to decipher computational frameworks modeling operations of HCRT networks.


Subject(s)
Arousal/physiology , Orexin Receptors/physiology , Orexins/physiology , Sleep/physiology , Wakefulness/physiology , Animals , History, 20th Century , History, 21st Century , Humans , Orexins/history
13.
Front Neurol Neurosci ; 45: 38-51, 2021.
Article in English | MEDLINE | ID: mdl-34052810

ABSTRACT

Orexins regulate a wide variety of biological functions, most notably the sleep-wake cycle, reward and stress processing, alertness, vigilance, and cognitive functioning. Alterations of central and peripheral orexin levels are linked to conditions such as narcolepsy, anorexia nervosa, age-related cognitive decline, and neurodegenerative disease. Preliminary studies suggest that orexin mimetics can safely promote the wake signal via orexin agonism during the day and that orexin receptor antagonists can promote the sleep signal during the night. Thus, novel orexin therapies have the potential to either improve memory, cognition, and daytime performance directly or indirectly, through promotion of good sleep. The full scope of the therapeutic potential of orexin therapies remains to be elucidated.


Subject(s)
Aging/metabolism , Alzheimer Disease/drug therapy , Anorexia Nervosa/metabolism , Cognitive Dysfunction/drug therapy , Narcolepsy/drug therapy , Orexin Receptors/drug effects , Orexins/physiology , Parkinson Disease/drug therapy , Sleep/physiology , Alzheimer Disease/metabolism , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Humans , Narcolepsy/complications , Orexins/metabolism , Parkinson Disease/metabolism
14.
Front Neurol Neurosci ; 45: 22-37, 2021.
Article in English | MEDLINE | ID: mdl-34052813

ABSTRACT

The hypocretins/orexins are two excitatory neuropeptides, alternately called HCRT1 or orexin-A and HCRT2 or orexin-B, that are the endogenous ligands for two G-protein-coupled receptors, HCRTR1/OX1R and HCRTR2/OX2R. Shortly after the discovery of this system, degeneration of hypocretin/orexin-producing neurons was implicated in the etiology of the sleep disorder narcolepsy. The involvement of this system in a disorder characterized by the loss of control over arousal state boundaries also suggested its role as a critical component of endogenous sleep-wake regulatory circuitry. The broad projections of the hypocretin/orexin-producing neurons, along with differential expression of the two receptors in the projection fields of these neurons, suggest distinct roles for these receptors. While HCRTR1/OX1R is associated with regulation of motivation, reward, and autonomic functions, HCRTR2/OX2R is strongly linked to sleep-wake control. The association of hypocretin/orexin with these physiological processes has led to intense interest in the therapeutic potential of compounds targeting these receptors. Agonists and antagonists for the hypocretin/orexin receptors have shown potential for the treatment of disorders of excessive daytime somnolence and nocturnal hyperarousal, respectively, with the first antagonists approved by the US Food and Drug Administration (FDA) in 2014 and 2019 for the treatment of insomnia. These and related compounds have also been useful tools to advance hypocretin/orexin neurobiology.


Subject(s)
Disorders of Excessive Somnolence/drug therapy , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/agonists , Orexin Receptors/physiology , Orexins/physiology , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep Wake Disorders/drug therapy , Sleep/physiology , Wakefulness/physiology , Animals , Humans , Sleep/drug effects , Wakefulness/drug effects
15.
Front Neurol Neurosci ; 45: 61-74, 2021.
Article in English | MEDLINE | ID: mdl-34052814

ABSTRACT

The multifunctional, hypothalamic hypocretin/orexin (HCRT)-producing neurons regulate an array of physiological and behavioral states including arousal, sleep, feeding, emotions, stress, and reward. How a presumably uniform HCRT neuron population regulates such a diverse set of functions is not clear. The role of the HCRT neuropeptides may vary depending on the timing and localization of secretion and neuronal activity. Moreover, HCRT neuropeptides may not mediate all functions ascribed to HCRT neurons. Some could be orchestrated by additional neurotransmitters and neuropeptides that are expressed in HCRT neurons. We hypothesize that HCRT neurons are segregated into genetically, anatomically and functionally distinct subpopulations. We discuss accumulating data that suggest the existence of such HCRT neuron subpopulations that may effectuate the diverse functions of these neurons in mammals and fish.


Subject(s)
Nerve Net/physiology , Neurons/physiology , Orexins/physiology , Animals , Humans , Nerve Net/metabolism , Neurons/classification , Neurons/metabolism , Orexins/metabolism
16.
Front Neurol Neurosci ; 45: 75-90, 2021.
Article in English | MEDLINE | ID: mdl-34052816

ABSTRACT

During the last decade, optogenetic-based circuit mapping has become one of the most common approaches to systems neuroscience, and amassing studies have expanded our understanding of brain structures causally involved in the regulation of sleep-wake cycles. Recent imaging technologies enable the functional mapping of cellular activity, from population down to single-cell resolution, across a broad repertoire of behaviors and physiological processes, including sleep-wake states. This chapter summarizes experimental evidence implicating hypocretins/orexins, melanin-concentrating hormone, and inhibitory neurons from the lateral hypothalamus (LH) in forming an intricate network involved in regulating sleep and metabolism, including feeding behaviors. It further confirms the dual sleep-metabolic functions of LH cells, and sheds light on a possible mechanism underlying brain plasticity during sleep and metabolic disorders.


Subject(s)
Feeding Behavior/physiology , Hypothalamic Area, Lateral/physiology , Hypothalamic Hormones/physiology , Melanins/physiology , Nerve Net/physiology , Neurons/physiology , Orexins/physiology , Pituitary Hormones/physiology , Sleep/physiology , Animals , Humans , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Melanins/metabolism , Nerve Net/metabolism , Neurons/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism
18.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925368

ABSTRACT

Orexin is expressed mainly in the hypothalamus and is known to activate the hypothalamic-pituitary-adrenal (HPA) axis that is involved in various stress responses and its resilience. However, the effects of orexin on the endocrine function of pituitary corticotrope cells remain unclear. In this study, we investigated the roles of orexin A in pro-opiomelanocortin (POMC) transcription using mouse corticotrope AtT20 cells, focusing on the bone morphogenetic protein (BMP) system expressed in the pituitary. Regarding the receptors for orexin, type 2 (OXR2) rather than type 1 (OX1R) receptor mRNA was predominantly expressed in AtT20 cells. It was found that orexin A treatment enhanced POMC expression, induced by corticotropin-releasing hormone (CRH) stimulation through upregulation of CRH receptor type-1 (CRHR1). Orexin A had no direct effect on the POMC transcription suppressed by BMP-4 treatment, whereas it suppressed Smad1/5/9 phosphorylation and Id-1 mRNA expression induced by BMP-4. It was further revealed that orexin A had no significant effect on the expression levels of type I and II BMP receptors but upregulated inhibitory Smad6/7 mRNA and protein levels in AtT20 cells. The results demonstrated that orexin A upregulated CRHR signaling and downregulated BMP-Smad signaling, leading to an enhancement of POMC transcription by corticotrope cells.


Subject(s)
Orexins/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Corticotrophs/metabolism , Corticotropin-Releasing Hormone/metabolism , Mice , Orexins/physiology , Phosphorylation , Pituitary Gland/metabolism , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects
19.
Peptides ; 139: 170530, 2021 05.
Article in English | MEDLINE | ID: mdl-33741478

ABSTRACT

Our daily life does not only involve a calm resting state but is rather full of perturbations that induce active states such as moving, eating, and communicating. During such active conditions, cardiorespiratory regulation should be adjusted according to bodily demand, which differs from that during the resting state, by modulating or resetting the operating point. To explore neural mechanisms in the state-dependent adjustment of central autonomic regulation, my research group has recently focused on the fight-or-flight response because the stressor induces not only cognitive, emotional, and behavioral changes but also autonomic changes. In this brief review, I will summarize our discovery using orexin knockout mice and orexin neuron-ablated mice for the possible contribution of orexin, a hypothalamic neuropeptide, to the state-dependent adjustment of the central autonomic regulation. In addition, I will introduce some recent discovery using optogenetic manipulation of the orexin and related systems. The diversity of synaptic control of the cardiovascular and respiratory neurons appears necessary for animals to adapt themselves to ever-changing life circumstances and behavioral states. The orexin system is likely to function as one of the essential modulators for coordinating the circuits controlling autonomic functions and behaviors.


Subject(s)
Autonomic Nervous System/physiology , Orexins/physiology , Stress, Physiological/physiology , Animals , Homeostasis , Mice , Mice, Transgenic , Optogenetics
20.
Dis Model Mech ; 14(1)2021 01 26.
Article in English | MEDLINE | ID: mdl-33735098

ABSTRACT

The development of animal models is a critical step for exploring the underlying pathophysiological mechanisms of major affective disorders and for evaluating potential therapeutic approaches. Although most neuropsychiatric research is performed on nocturnal rodents, differences in how diurnal and nocturnal animals respond to changing photoperiods, combined with a possible link between circadian rhythm disruption and affective disorders, has led to a call for the development of diurnal animal models. The need for diurnal models is most clear for seasonal affective disorder (SAD), a widespread recurrent depressive disorder that is linked to exposure to short photoperiods. Here, we briefly review what is known regarding the etiology of SAD and then examine progress in developing appropriate diurnal rodent models. Although circadian disruption is often invoked as a key contributor to SAD, a mechanistic understanding of how misalignment between endogenous circadian physiology and daily environmental rhythms affects mood is lacking. Diurnal rodents show promise as models of SAD, as changes in affective-like behaviors are induced in response to short photoperiods or dim-light conditions, and symptoms can be ameliorated by brief exposure to intervals of bright light coincident with activity onset. One exciting avenue of research involves the orexinergic system, which regulates functions that are disturbed in SAD, including sleep cycles, the reward system, feeding behavior, monoaminergic neurotransmission and hippocampal neurogenesis. However, although diurnal models make intuitive sense for the study of SAD and are more likely to mimic circadian disruption, their utility is currently hampered by a lack of genomic resources needed for the molecular interrogation of potential mechanisms.


Subject(s)
Circadian Rhythm , Depression/physiopathology , Light , Photoperiod , Seasonal Affective Disorder/physiopathology , Animals , Anxiety/psychology , Behavior, Animal/physiology , Choice Behavior , Cognition , Disease Models, Animal , Humans , Maze Learning , Orexins/physiology , Social Behavior , Swimming/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...