1
|
Dovek L, Tinsley CE, Gutowsky K, McDaniel K, Potter Z, Ruffins M, Milman NEP, Wong C, Soumyanath A, Gray NE, Lim MM. Centella asiatica improves sleep quality and quantity in aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636070. [PMID: 39974941 PMCID: PMC11838466 DOI: 10.1101/2025.02.01.636070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Age-related sleep disruption is common in older adults. Not only does the total amount of time spent in sleep decline, but the number of arousals during sleep increases with age. As sleep is important for both memory consolidation and to prevent neurodegenerative pathology, this decline in sleep and/or sleep consolidation may underlie age-related cognitive decline and dementias. Furthermore, treatment of sleep disruption can improve quality of life. However, few interventions have successfully reversed age-related sleep decline. Extracts from the plant Centella asiatica have demonstrated neuroprotective effects in human, rodent, and fly models of aging and neurodegenerative diseases, and is a promising intervention for dementias, yet little is known about how these extracts affect sleep patterns. Here, we administered Centella asiatica water extract ( CAW) dosed or control chow to male and female C57BL6/J mice aged 18 months. Effects on sleep composition were determined using electrodes that recorded EEG and EMG signals. We found that CAW dosed chow (1000 mg/kg/day) increased REM sleep time in aged male mice and decreased the number of arousals during sleep observed in aged females, compared to age- and sex-matched controls. We conclude that CAW administered in food has a moderate, sex-dependent effect on sleep quantity and quality. Statement of Significance Sleep declines with age and may underline age-related cognitive changes. However, few interventions have successfully reversed age-related sleep and cognitive decline. This study found that botanical extract from the plant Centella asiatica increased total REM sleep time in aged male mice, and decreased sleep fragmentation in aged female mice, compared to age- and sex-matched controls. Whether these moderate, sex-dependent effect sizes on sleep in aged mice are impactful enough to affect cognition, quality of life, and/or neurodegenerative pathology could be explored in future studies.
Collapse
|
2
|
Tóth A, Dobolyi Á. Prolactin in sleep and EEG regulation: New mechanisms and sleep-related brain targets complement classical data. Neurosci Biobehav Rev 2025; 169:106000. [PMID: 39755290 DOI: 10.1016/j.neubiorev.2024.106000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
The role of prolactin in sleep regulation has been the subject of extensive research over the past 50 years, resulting in the identification of multiple, disparate functions for the hormone. Prolactin demonstrated a characteristic circadian release pattern with elevation during dark and diminution during light. High prolactin levels were linked to non-rapid eye movement sleep and electroencephalogram delta activity in humans. Conversely, hyperprolactinemia showed strong correlation with REM sleep in rodent studies. Prolactin may be implicated in the alterations in female sleep patterns observed during the reproductive cycle, it may play a role in the REM sleep enhancement following stress and in sleep-related immunological processes. In conclusion, prolactin appears to have a sleep-promoting role, particularly during the dark phase. However, it does not appear to play a central and coherent role in sleep regulation, as observed in some neuropeptides such as orexin. Conversely, its principal function may be to facilitate situational, yet adaptive, changes in sleep patterns in response to challenging physiological phases, such as those associated with stress, immunological challenges, or the reproductive cycle. Neuronal substrates for prolactin-mediated sleep effects remain unknown; however, recent rodent sleep studies may provide insights into the potential sites of these effects.
Collapse
Affiliation(s)
- Attila Tóth
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Hungary.
| | - Árpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Hungary
| |
Collapse
|
3
|
Kaur M, Mehta R, Muthuswami R, Mallick BN. Noradrenaline enhances Na-K ATPase subunit expression by HuR-induced mRNA stabilization and their transportation to the cell surface through PLC and PKC mediated pathway: Implications with REMS-loss associated disorders. J Neurochem 2024; 168:2561-2576. [PMID: 38676340 DOI: 10.1111/jnc.16116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/08/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Rapid eye movement sleep (REMS) maintains brain excitability at least by regulating Na-K ATPase activity. Although REMS deprivation (REMSD)-associated elevated noradrenaline (NA) increases Na-K ATPase protein expression, its mRNA transcription did not increase. We hypothesized and confirmed both in vivo as well as in vitro that elevated mRNA stability explains the apparent puzzle. The mRNA stability was measured in control and REMSD rat brain with or without in vivo treatment with α1-adrenoceptor (AR) antagonist, prazosin (PRZ). Upon REMSD, Na-K ATPase α1-, and α2-mRNA stability increased significantly, which was prevented by PRZ. To decipher the molecular mechanism of action, we estimated NA-induced Na-K ATPase mRNA stability in Neuro-2a cells under controlled conditions and by transcription blockage using Actinomycin D (Act-D). NA increased Na-K ATPase mRNA stability, which was prevented by PRZ and propranolol (PRP, β-AR antagonist). The knockdown assay confirmed that the increased mRNA stabilization was induced by elevated cytoplasmic abundance of Human antigen R (HuR) and involving (Phospholipase C) PLC-mediated activation of Protein Kinase C (PKC). Additionally, using cell-impermeable Enz-link sulfo NHS-SS-Biotin, we observed that NA increased Na-K ATPase α1-subunits on the Neuro-2a cell surface. We conclude that REMSD-associated elevated NA, acting on α1- and β-AR, increases nucleocytoplasmic translocation of HuR and increases Na-K ATPase mRNA stability, resulting in increased Na-K ATPase protein expression. The latter then gets translocated to the neuronal membrane surface involving both PKC and (Protein Kinase A) PKA-mediated pathways. These findings may be exploited for the amelioration of REMSD-associated chronic disorders and symptoms.
Collapse
Affiliation(s)
- Manjeet Kaur
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rachna Mehta
- AMITY Institute of Neuropsychology and Neurosciences, AMITY University Uttar Pradesh, Noida, UP, India
| | - Rohini Muthuswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Birendra Nath Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- AMITY Institute of Neuropsychology and Neurosciences, AMITY University Uttar Pradesh, Noida, UP, India
| |
Collapse
|
4
|
Maurer JJ, Lin A, Jin X, Hong J, Sathi N, Cardis R, Osorio-Forero A, Lüthi A, Weber F, Chung S. Homeostatic regulation of rapid eye movement sleep by the preoptic area of the hypothalamus. eLife 2024; 12:RP92095. [PMID: 38884573 PMCID: PMC11182646 DOI: 10.7554/elife.92095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Abstract
Rapid eye movement sleep (REMs) is characterized by activated electroencephalogram (EEG) and muscle atonia, accompanied by vivid dreams. REMs is homeostatically regulated, ensuring that any loss of REMs is compensated by a subsequent increase in its amount. However, the neural mechanisms underlying the homeostatic control of REMs are largely unknown. Here, we show that GABAergic neurons in the preoptic area of the hypothalamus projecting to the tuberomammillary nucleus (POAGAD2→TMN neurons) are crucial for the homeostatic regulation of REMs in mice. POAGAD2→TMN neurons are most active during REMs, and inhibiting them specifically decreases REMs. REMs restriction leads to an increased number and amplitude of calcium transients in POAGAD2→TMN neurons, reflecting the accumulation of REMs pressure. Inhibiting POAGAD2→TMN neurons during REMs restriction blocked the subsequent rebound of REMs. Our findings reveal a hypothalamic circuit whose activity mirrors the buildup of homeostatic REMs pressure during restriction and that is required for the ensuing rebound in REMs.
Collapse
Affiliation(s)
- John J Maurer
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Alexandra Lin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Xi Jin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jiso Hong
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas Sathi
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Romain Cardis
- Department of Fundamental Neurosciences, University of LausanneLausanneSwitzerland
| | | | - Anita Lüthi
- Department of Fundamental Neurosciences, University of LausanneLausanneSwitzerland
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
5
|
Manoharan N, Parasuraman R, Jayamurali D, Muthusamy P, Govindarajulu S. Role of Thymoquinone on sleep restriction and its mitigating effect on leptin-mediated signaling pathway in rat brain. Mol Biol Rep 2024; 51:769. [PMID: 38886257 DOI: 10.1007/s11033-024-09699-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Sleep and stress interact bidirectionally by acting on brain circuits that affect metabolism. Sleep and its alterations have impact on blood leptin levels, metabolic hormone that regulates appetite. Brain expresses the receptors for the peptide hormone leptin produced from adipocytes. The hypothalamic orexin neurons are low during sleep and active when awake, influenced by a complex interaction with leptin. Thymoquinone was found to be the major bioactive component of Nigella sativa. The aim of this study was to study the role of thymoquinone on sleep restriction and its mitigating effect on leptin-mediated signaling pathway in rat brain. METHODS AND RESULTS 30 adult male Wistar rats were divided into 5 groups with 6 animals in each group: Control; Thymoquinone (TQ); Corn oil; Chronic Sleep restriction (CSR); and CSR + TQ. After 30 days, behavioral analysis, antioxidant, lipid profile, glucose level, liver and kidney function test, neurotransmitters, neuropeptides, and mRNA expression in in vivo studies were also assessed and pharmacokinetic and docking were done for thymoquinone. Thymoquinone has also shown good binding affinity to the target proteins. CSR has induced oxidative stress in the discrete brain regions and plasma. Current study has shown many evidences that sleep restriction has altered the neurobehavioral, antioxidant status, lipid profile, neurotransmitters, neuropeptide levels, and feeding behavior which damage the Orexin-leptin system which regulates the sleep and feeding that leads to metabolic dysfunction. CONCLUSION The potentiality of Thymoquinone was revealed in in silico studies, and its action in in vivo studies has proved its effectiveness. The study concludes that Thymoquinone has exhibited its effect by diminishing the metabolic dysfunction by its neuroprotective, antioxidant, and hypolipidemic properties.
Collapse
Affiliation(s)
- Nivedita Manoharan
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Rajeshwari Parasuraman
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Dheepthi Jayamurali
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Pazhanisankar Muthusamy
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India
| | - Sathyanarayanan Govindarajulu
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600 113, India.
| |
Collapse
|
6
|
Marquezini BP, Moysés-Oliveira M, Kloster A, Cunha L, Deconto TB, Mosini AC, Guerreiro P, Paschalidis M, Adami LNG, Andersen ML, Tufik S. Exploring the molecular pathways linking sleep phenotypes and POGZ-associated neurodevelopmental disorder. J Med Genet 2024; 61:586-589. [PMID: 38350721 DOI: 10.1136/jmg-2023-109508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024]
Abstract
Pogo transposable element-derived protein with ZNF domain (POGZ) gene encodes a chromatin regulator and rare variants on this gene have been associated with a broad spectrum of neurodevelopmental disorders, such as White-Sutton syndrome. Patient clinical manifestations frequently include developmental delay, autism spectrum disorder and obesity. Sleep disturbances are also commonly observed in these patients, yet the biological pathways which link sleep traits to the POGZ-associated syndrome remain unclear. We screened for sleep implications among individuals with causative POGZ variants previously described. Sleep disturbances were observed in 52% of patients, and being obese was not observed as a risk factor for sleep problems. Next, we identified genes associated with sleep-associated traits among the POGZ regulatory targets, aiming to uncover the molecular pathways that, when disrupted by POGZ loss of function, contribute to the aetiology of sleep phenotypes in these patients. The intersect between POGZ targets and sleep-related genes was used in a pathway enrichment analysis. Relevant pathways among these overlapping genes are involved in the regulation of circadian rhythm, tau protein binding, ATPase activator activity. This study may represent the beginning for novel functional investigations on shared molecular mechanisms between sleep disturbances and rare developmental syndromes related to POGZ and its regulatory targets.
Collapse
Affiliation(s)
| | | | - Anna Kloster
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | - Lais Cunha
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | | | | | - Pedro Guerreiro
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | - Mayara Paschalidis
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | | | - Monica Levy Andersen
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Dugan BJ, Fraigne JJ, Peever J. REM sleep: Out-dreaming fear. Curr Biol 2024; 34:R510-R512. [PMID: 38772341 DOI: 10.1016/j.cub.2024.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
The ability to forget fear-inducing situations is essential for adapting to our environment, but the neural mechanisms underlying 'fear forgetting' remain unclear. Novel findings reveal that the activity of the infralimbic cortex - specifically during REM sleep - contributes to the extinction of fear memory.
Collapse
Affiliation(s)
- Brittany J Dugan
- Department of Cell and Systems Biology, University of Toronto, Toronto ON, Canada
| | - Jimmy J Fraigne
- Department of Cell and Systems Biology, University of Toronto, Toronto ON, Canada
| | - John Peever
- Department of Cell and Systems Biology, University of Toronto, Toronto ON, Canada.
| |
Collapse
|
8
|
Hong J, Choi K, Fuccillo MV, Chung S, Weber F. Infralimbic activity during REM sleep facilitates fear extinction memory. Curr Biol 2024; 34:2247-2255.e5. [PMID: 38714199 PMCID: PMC11111341 DOI: 10.1016/j.cub.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 05/09/2024]
Abstract
Rapid eye movement (REM) sleep is known to facilitate fear extinction and play a protective role against fearful memories.1,2 Consequently, disruption of REM sleep after a traumatic event may increase the risk for developing PTSD.3,4 However, the underlying mechanisms by which REM sleep promotes extinction of aversive memories remain largely unknown. The infralimbic cortex (IL) is a key brain structure for the consolidation of extinction memory.5 Using calcium imaging, we found in mice that most IL pyramidal neurons are intensively activated during REM sleep. Optogenetically suppressing the IL specifically during REM sleep within a 4-h window after auditory-cued fear conditioning impaired extinction memory consolidation. In contrast, REM-specific IL inhibition after extinction learning did not affect the extinction memory. Whole-cell patch-clamp recordings demonstrated that inactivating IL neurons during REM sleep depresses their excitability. Together, our findings suggest that REM sleep after fear conditioning facilitates fear extinction by enhancing IL excitability and highlight the importance of REM sleep in the aftermath of traumatic events for protecting against traumatic memories.
Collapse
Affiliation(s)
- Jiso Hong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyuhyun Choi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc V Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinjae Chung
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Franz Weber
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Abhishek K, Mallick BN. Sleep loss disrupts decision-making ability and neuronal cytomorphology in zebrafish and the effects are mediated by noradrenaline acting on α1-adrenoceptor. Neuropharmacology 2024; 247:109861. [PMID: 38331315 DOI: 10.1016/j.neuropharm.2024.109861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/21/2023] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
Sleep is an instinct behavior, and its significance and functions are still an enigma. It is expressed throughout one's life and its loss affects psycho-somatic and physiological processes. We had proposed that it might maintain a fundamental property of the neurons and the brain. In that context, it was shown that sleep, rapid eye movement sleep (REMS) in particular, by regulating noradrenaline (NA), maintains the brain excitability. It was also reported that sleep-loss affected memory, reaction time and decision-making ability among others. However, as there was lack of clarity on the cause-and-effect relationship as to how the sleep-loss could affect these basic behaviors, their association was questioned and it was difficult to propose a cure or at least ways and means to ameliorate the symptoms. Also, we wanted to conduct the studies in a simpler model system so that conducting future molecular studies might be easier. Hence, using zebrafish as a model we evaluated if sleep-loss affected the basic decision-making ability, a cognitive process and if the effect was induced by NA. Indeed, our findings confirmed that upon sleep-deprivation, the cognitive decision-making ability of the prey zebrafish was compromised to protect itself by running away from the reach of the exposed predator Tiger Oscar (TO) fish. Also, we observed that upon sleep-loss the axonal arborization of the prey zebrafish brain was reduced. Interestingly, the effects were prevented by prazosin (PRZ), an α1-adrenoceptor (AR) antagonist and when the zebrafish recovered from the lost sleep.
Collapse
Affiliation(s)
- Kumar Abhishek
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Birendra Nath Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Amity Institute of Neuropsychology & Neurosciences, Amity University Uttar Pradesh, Sector 125, NOIDA, 201313, India.
| |
Collapse
|
10
|
Sardi NF, Pescador AC, Azevedo EM, Pochapski JA, Kukolj C, Spercoski KM, Andrade AJM, da Cunha C, Fischer L. Sleep and Pain: A Role for the Anterior Cingulate Cortex, Nucleus Accumbens, and Dopamine in the Increased Pain Sensitivity Following Sleep Restriction. THE JOURNAL OF PAIN 2024; 25:331-349. [PMID: 37673193 DOI: 10.1016/j.jpain.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Persistent pain conditions and sleep disorders are public health problems worldwide. It is widely accepted that sleep disruption increases pain sensitivity; however, the underlying mechanisms are poorly understood. In this study, we used a protocol of 6 hours a day of total sleep deprivation for 3 days in rats to advance the understanding of these mechanisms. We focused on gender differences and the dopaminergic mesocorticolimbic system. The findings demonstrated that sleep restriction (SR) increased pain sensitivity in a similar way in males and females, without inducing a significant stress response. This pronociceptive effect depends on a nucleus accumbens (NAc) neuronal ensemble recruited during SR and on the integrity of the anterior cingulate cortex (ACC). Data on indirect dopaminergic parameters, dopamine transporter glycosylation, and dopamine and cyclic adenosine monophosphate (AMP)-regulated phosphoprotein-32 phosphorylation, as well as dopamine, serotonin, and norepinephrine levels, suggest that dopaminergic function decreases in the NAc and ACC after SR. Complementarily, pharmacological activation of dopamine D2, but not D1 receptors either in the ACC or in the NAc prevents SR from increasing pain sensitivity. The ACC and NAc are the main targets of dopaminergic mesocorticolimbic projections with a key role in pain modulation. This study showed their integrative role in the pronociceptive effect of SR, pointing to dopamine D2 receptors as a potential target for pain management in patients with sleep disorders. These findings narrow the focus of future studies on the mechanisms by which sleep impairment increases pain sensitivity. PERSPECTIVE: This study demonstrates that the pronociceptive effect of SR affects similarly males and females and depends on a NAc neuronal ensemble recruited during SR and on the integrity of the ACC. Findings on dopaminergic function support dopamine D2 receptors as targets for pain management in sleep disorders patients.
Collapse
Affiliation(s)
- Natalia F Sardi
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Ana C Pescador
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Evellyn M Azevedo
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - José A Pochapski
- Department of Pharmacology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil; Department of Biochemistry, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Caroline Kukolj
- Department of Biochemistry, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Katherinne M Spercoski
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil; Division of Biosciences, Federal University of Parana, Palotina, Parana, Brazil
| | - Anderson J M Andrade
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Claudio da Cunha
- Department of Pharmacology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Luana Fischer
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| |
Collapse
|
11
|
Giri S, Mehta R, Mallick BN. REM Sleep Loss-Induced Elevated Noradrenaline Plays a Significant Role in Neurodegeneration: Synthesis of Findings to Propose a Possible Mechanism of Action from Molecule to Patho-Physiological Changes. Brain Sci 2023; 14:8. [PMID: 38275513 PMCID: PMC10813190 DOI: 10.3390/brainsci14010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
Wear and tear are natural processes for all living and non-living bodies. All living cells and organisms are metabolically active to generate energy for their routine needs, including for survival. In the process, the cells are exposed to oxidative load, metabolic waste, and bye-products. In an organ, the living non-neuronal cells divide and replenish the lost or damaged cells; however, as neuronal cells normally do not divide, they need special feature(s) for their protection, survival, and sustenance for normal functioning of the brain. The neurons grow and branch as axons and dendrites, which contribute to the formation of synapses with near and far neurons, the basic scaffold for complex brain functions. It is necessary that one or more basic and instinct physiological process(es) (functions) is likely to contribute to the protection of the neurons and maintenance of the synapses. It is known that rapid eye movement sleep (REMS), an autonomic instinct behavior, maintains brain functioning including learning and memory and its loss causes dysfunctions. In this review we correlate the role of REMS and its loss in synaptogenesis, memory consolidation, and neuronal degeneration. Further, as a mechanism of action, we will show that REMS maintains noradrenaline (NA) at a low level, which protects neurons from oxidative damage and maintains neuronal growth and synaptogenesis. However, upon REMS loss, the level of NA increases, which withdraws protection and causes apoptosis and loss of synapses and neurons. We propose that the latter possibly causes REMS loss associated neurodegenerative diseases and associated symptoms.
Collapse
Affiliation(s)
- Shatrunjai Giri
- Department of Biosciences, Manipal University Jaipur, Jaipur 303007, India;
| | - Rachna Mehta
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida 201301, India;
| | - Birendra Nath Mallick
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida 201301, India;
| |
Collapse
|
12
|
Ratna D, Mondal AC, Mallick BN. Modulation of dopamine from ventral tegmental area neurons by the LC-REM-OFF and PPT-REM-ON neurons in REMS regulation in freely moving rats. Neuropharmacology 2023:109621. [PMID: 37276957 DOI: 10.1016/j.neuropharm.2023.109621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/07/2023]
Abstract
The role of dopamine (DA)-ergic neurons in ventral tegmental area (VTA) in schizophrenia, depression, hallucinations have been extensively studied. Rapid eye movement sleep (REMS), the closest objective correlate of dream and hallucination, is disrupted during these psychological dysfunctions; however, it was unknown if there is any common neuronal substrate for their regulation. Interactions among locus coeruleus (LC) REM-OFF and pedunculopontine tegmentum (PPT) REM-ON neurons have been reported to regulate REMS in health and diseases. Recently we have reported that PPT neurons modulate VTA and REMS. However, although VTA-DA neurons receive projections from LC and PPT, their role in REMS regulation was unclear. We proposed that the LC and PPT might intermittently modulate VTA-DA neurons and modulate REMS. Male Wistar rats were surgically prepared and electrophysiological wakefulness-sleep-REMS recorded in chronic freely moving condition. We employed RNAi induced downregulation of tyrosine hydroxylase (TH) to evaluate the role of VTA-DA in regulating REMS. We observed that TH-knockdown in VTA decreased REMS in experimental rats, which returned to baseline upon PPT stimulation. Thus, VTA-DA neurons are activated by the REM-ON neurons to modulate REMS, the closest objectively recordable correlate of dreams. In these animals, LC stimulation altered Non-REMS and waking. Based on the findings we have discussed the role of VTA neurochemical circuitry in REMS regulation and their possible implications with REMS-associated dreaming and hallucination in health and diseases.
Collapse
Affiliation(s)
- Deshdeepak Ratna
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Birendra Nath Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Amity Institute of Neuropsychology & Neurosciences, Amity University Campus, Sector 125, Noida, 201313, Gautam Budh Nagar, Uttar Pradesh, India.
| |
Collapse
|
13
|
Sleep and Neuroimmunomodulation for Maintenance of Optimum Brain Function: Role of Noradrenaline. Brain Sci 2022; 12:brainsci12121725. [PMID: 36552184 PMCID: PMC9776456 DOI: 10.3390/brainsci12121725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Immune function and sleep are two normal physiological processes to protect the living organism from falling sick. There is hardly any disease in which they remain unaffected, though the quantum of effect may differ. Therefore, we propose the existence of a strong correlation between sleep (quality or quantity) and immune response. This may be supported by the fact that sleep loss modulates many of the immunological molecules, which includes interferons; however, not much is known about their mechanism of action. Sleep is divided into rapid eye movement sleep (REMS) and non-REMS. For practical reasons, experimental studies have been conducted mostly by inducing loss of REMS. It has been shown that withdrawal of noradrenaline (NA) is a necessity for generation of REMS. Moreover, NA level increases in the brain upon REMS loss and the elevated NA is responsible for many of the sleep loss-associated symptoms. In this review, we describe how sleep (and its disturbance/loss) modulates the immune system by modulating the NA level in the brain or vice versa to maintain immune functions, physiological homeostasis, and normal healthy living. The increased levels of NA during REMS loss may cause neuroinflammation possibly by glial activation (as NA is a key modulator of microglia). Therefore, maintaining sleep hygiene plays a crucial role for a normal healthy living.
Collapse
|
14
|
Wofford N, Ceballos N, Elkins G, Westerberg CE. A brief nap during an acute stressor improves negative affect. J Sleep Res 2022; 31:e13701. [PMID: 35851731 PMCID: PMC9786543 DOI: 10.1111/jsr.13701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/30/2022]
Abstract
Overnight sleep can reduce perceived stress, and improve associated cognitive disruptions and negative affect after an acute stressor. Whether a brief nap can also bestow these benefits in a non-sleep-restricted population is currently unknown. In this study that used a between-subjects design, stress was triggered by administering a modified Trier Social Stress Test to two groups of participants (nap [n = 29], wake [n = 41]). All participants were instructed they would give a speech during the study but the topic would be withheld until later, and then completed a math task. After a 40-min break in which participants watched a neutral video or took a nap monitored with electroencephalography, stress was reinforced by presenting the speech topics and giving participants a 10-min preparation period. Next, instead of giving a speech, the study ended and participants were debriefed. Negative affect, perceived stress and working memory were measured at multiple time points before and after the break. Both groups showed lower perceived stress and improved working memory after the break than before, but a nap did not confer additional benefits for perceived stress or working memory beyond taking a break. However, the nap group exhibited lower negative affect after the break than the wake group, and only the nap group showed a reduction in negative affect compared with initial negative affect levels. These results indicate a nap can improve negative emotions accompanying a stressor to a greater extent than taking a break, and suggest that brief naps may be a useful way to improve mood while experiencing an acute stressor.
Collapse
Affiliation(s)
- Nathan Wofford
- Department of PsychologyTexas State UniversitySan MarcosTexasUSA,Department of Psychology and NeuroscienceBaylor UniversityWacoTexasUSA
| | - Natalie Ceballos
- Department of PsychologyTexas State UniversitySan MarcosTexasUSA
| | - Gary Elkins
- Department of Psychology and NeuroscienceBaylor UniversityWacoTexasUSA
| | | |
Collapse
|
15
|
López-Muciño LA, García-García F, Cueto-Escobedo J, Acosta-Hernández M, Venebra-Muñoz A, Rodríguez-Alba JC. Sleep loss and addiction. Neurosci Biobehav Rev 2022; 141:104832. [PMID: 35988803 DOI: 10.1016/j.neubiorev.2022.104832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
Reducing sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric disorders. Furthermore, previous studies have shown that reduction in sleep time is a factor that favors relapse in addicted patients. Additionally, animal models have demonstrated that both sleep restriction and sleep deprivation increase the preference for alcohol, methylphenidate, and the self-administration of cocaine. Therefore, the present review discusses current knowledge about the influence of sleep hours reduction on addictivebehaviors; likewise, we discuss the neuronal basis underlying the sleep reduction-addiction relationship, like the role of the orexin and dopaminergic system and neuronal plasticity (i.e., delta FosB expression). Potentially, chronic sleep restriction could increase brain vulnerability and promote addictive behavior.
Collapse
Affiliation(s)
- Luis Angel López-Muciño
- Health Sciences Ph.D. Program, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Fabio García-García
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Jonathan Cueto-Escobedo
- Department of Clinical and Translational Research, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Mario Acosta-Hernández
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Arturo Venebra-Muñoz
- Laboratory of Neurobiology of Addiction and Brain Plasticity, Faculty of Science, Autonomous University of Mexico State, Edomex 50295, Mexico.
| | - Juan Carlos Rodríguez-Alba
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| |
Collapse
|
16
|
Niu L, Zhang F, Xu X, Yang Y, Li S, Liu H, Le W. Chronic sleep deprivation altered the expression of circadian clock genes and aggravated Alzheimer's disease neuropathology. Brain Pathol 2021; 32:e13028. [PMID: 34668266 PMCID: PMC9048513 DOI: 10.1111/bpa.13028] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/18/2021] [Accepted: 09/27/2021] [Indexed: 01/20/2023] Open
Abstract
Circadian disruption is prevalent in Alzheimer's disease (AD) and may contribute to cognitive impairment, psychological symptoms, and neurodegeneration. This study aimed to evaluate the effects of environmental and genetic factors on the molecular clock and to establish a link between circadian rhythm disturbance and AD. We investigated the pathological effects of chronic sleep deprivation (CSD) in the APPswe/PS1ΔE9 transgenic mice and their wild‐type (WT) littermates for 2 months and evaluated the expression levels of clock genes in the circadian rhythm‐related nuclei. Our results showed that CSD impaired learning and memory, and further exaggerated disease progression in the AD mice. Furthermore, CSD caused abnormal expression of Bmal1, Clock, and Cry1 in the circadian rhythm‐related nuclei of experimental mice, and these changes are more significant in AD mice. Abnormal expression of clock genes in AD mice suggested that the expression of clock genes is affected by APP/PS1 mutations. In addition, abnormal tau phosphorylation was found in the retrosplenial cortex, which was co‐located with the alteration of BMAL1 protein level. Moreover, the level of tyrosine hydroxylase in the locus coeruleus of AD and WT mice was significantly increased after CSD. There may be a potential link between the molecular clock, Aβ pathology, tauopathy, and the noradrenergic system. The results of this study provided new insights into the potential link between the disruption of circadian rhythm and the development of AD.
Collapse
Affiliation(s)
- Long Niu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Feng Zhang
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaojiao Xu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuting Yang
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hui Liu
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of Neurology and Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, Chengdu, China
| |
Collapse
|
17
|
Deng Y, Deng G, Grobe JL, Cui H. Hypothalamic GPCR Signaling Pathways in Cardiometabolic Control. Front Physiol 2021; 12:691226. [PMID: 34262481 PMCID: PMC8274634 DOI: 10.3389/fphys.2021.691226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/26/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is commonly associated with sympathetic overdrive, which is one of the major risk factors for the development of cardiovascular diseases, such as hypertension and heart failure. Over the past few decades, there has been a growing understanding of molecular mechanisms underlying obesity development with central origin; however, the relative contribution of these molecular changes to the regulation of cardiovascular function remains vague. A variety of G-protein coupled receptors (GPCRs) and their downstream signaling pathways activated in distinct hypothalamic neurons by different metabolic hormones, neuropeptides and monoamine neurotransmitters are crucial not only for the regulation of appetite and metabolic homeostasis but also for the sympathetic control of cardiovascular function. In this review, we will highlight the main GPCRs and associated hypothalamic nuclei that are important for both metabolic homeostasis and cardiovascular function. The potential downstream molecular mediators of these GPCRs will also be discussed.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- FOE Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
18
|
Giri S, Ranjan A, Kumar A, Amar M, Mallick BN. Rapid eye movement sleep deprivation impairs neuronal plasticity and reduces hippocampal neuronal arborization in male albino rats: Noradrenaline is involved in the process. J Neurosci Res 2021; 99:1815-1834. [PMID: 33819353 DOI: 10.1002/jnr.24838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 12/22/2022]
Abstract
Rapid eye movement sleep (REMS) favors brain development and memory, while it is decreased in neurodegenerative diseases. REMS deprivation (REMSD) affects several physiological processes including memory consolidation; however, its detailed mechanism(s) of action was unknown. REMS reduces, while REMSD elevates noradrenaline (NA) level in the brain; the latter induces several deficiencies and disorders, including changes in neuronal cytomorphology and apoptosis. Therefore, we proposed that REMS- and REMSD-associated modulation of NA level might affect neuronal plasticity and affect brain functions. Male albino rats were REMS deprived by flower-pot method for 6 days, and its effects were compared with home cage and large platform controls as well as post-REMSD recovered and REMS-deprived prazosin (α1-adrenoceptor antagonist)-treated rats. We observed that REMSD reduced CA1 and CA3 neuronal dendritic length, branching, arborization, and spine density, while length of active zone and expressions of pre- as well as post-synaptic proteins were increased as compared to controls; interestingly, prazosin prevented most of the effects in vivo. Studies on primary culture of neurons from chick embryo brain confirmed that NA at lower concentration(s) induced neuronal branching and arborization, while higher doses were destructive. The findings support our contention that REMSD adversely affects neuronal plasticity, branching, and synaptic scaffold, which explain the underlying cytoarchitectural basis of REMSD-associated patho-physio-behavioral changes. Consolidation of findings of this study along with that of our previous reports suggest that the neuronal disintegration could be due to either withdrawal of direct protective and proliferative role of low dose of NA or indirect effect of high dose of NA or both.
Collapse
Affiliation(s)
- Shatrunjai Giri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amit Ranjan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Awanish Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Megha Amar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
19
|
Tuan LH, Tsao CY, Lee LJH, Lee LJ. Voluntary exercise ameliorates synaptic pruning deficits in sleep-deprived adolescent mice. Brain Behav Immun 2021; 93:96-110. [PMID: 33358980 DOI: 10.1016/j.bbi.2020.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a critical period for brain development and adequate sleep during this period is essential for physical function and mental health. Emerging evidence has detailed the neurological impacts of sleep insufficiency on adolescents, as was unveiled by our previous study, microglia, one of the crucial contributors to synaptic pruning, is functionally disrupted by lack of sleep. Here, we provided evidence featuring the protective effect and the underlying mechanisms of voluntary exercise (VE) on microglial functions in an adolescent 72 h sleep deprivation (SD) model. We identified that the aberrant hippocampal neuronal activity and impaired short-term memory performance in sleep-deprived mice were prevented by 11 days of VE. VE significantly normalized the SD-induced dendritic spine increment and maintained the microglial phagocytic ability in sleep-deprived mice. Moreover, we found that the amendment of the noradrenergic signal in the central nervous system may explain the preventative effects of VE on the abnormalities of microglial and neuronal functions caused by SD. These data suggested that VE may confer protection to the microglia-mediated synaptic pruning in the sleep-deprived adolescent brains. Therefore, physical exercise could be a beneficial health practice for the adolescents that copes the adverse influence of inevitable sleep insufficiency.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Lukas Jyuhn-Hsiarn Lee
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
20
|
Flowerpot method for rapid eye movement sleep deprivation does not induce stress as defined by elevated serum corticosterone level in rats. Neurosci Lett 2021; 745:135631. [PMID: 33444674 DOI: 10.1016/j.neulet.2021.135631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 01/07/2023]
Abstract
Flowerpot method of rapid eye movement sleep (REMS) deprivation (REMSD) has been most extensively used in experiments to decipher the functions of REMS. The most common but serious criticism of this method has been presumed stress experienced by the experimental animals. The lack of systematic studies with appropriate controls to resolve this issue prompted this study. We have compared serum corticosterone levels as a marker of stress in male rats under REMSD by the flowerpot method and multiple types of control conditions. Additionally, to maintain consistency and uniformity of REMSD among groups, in the same rats, we estimated brain Na-K ATPase activity, which has been consistently reported to increase upon REMSD. The most effective method was one rat in single- or multiple-platforms set-up in a pool because it significantly increased Na-K ATPase activity without elevating serum corticosterone level. More than one rat in multiple platform set-up was ineffective and must be avoided. Also, large platform- and recovery-controls must be carried out simultaneously to rule out non-specific confounding effects.
Collapse
|
21
|
Shang FLT, Wanner SP, Damasceno WC, Martins YAT, Silva A, Prado LS. Independent effects of rapid eye movement sleep deprivation and exposure to environmental heat stress on aerobic performance and thermoregulatory responses in exercising rats. Temperature (Austin) 2020; 8:188-201. [PMID: 33997117 DOI: 10.1080/23328940.2020.1829939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Evidence indicates that aerobic performance is degraded either by environmental heat stress or sleep deprivation. However, whether these conditions interact to produce more significant performance impairment deserves further investigation. Therefore, this study investigated the effects of experimental sleep deprivation (24 h or 96 h) on aerobic performance and thermoregulatory responses in rats exercised on a treadmill at different environmental conditions. Adult male Wistar rats were subjected to rapid eye movement sleep deprivation (RSD) using the modified multiple platform method and were then subjected to an incremental-speed exercise until they were fatigued. Treadmill running was performed in a temperate (24°C) or warm (31°C) environment, and the colonic temperature (an index of core body temperature; TCORE) and the tail-skin temperature (TSKIN; an index of cutaneous heat loss) were recorded. 24-h and 96-h RSD produced small magnitude reductions in aerobic performance (Cohen's d = 0.47-0.58) and minor changes in thermoregulation. Relative to control rats, sleep-deprived rats showed a higher TCORE at the exercise initiation and a higher threshold for activating cutaneous heat loss, but unchanged TCORE and TSKIN at fatigue. Exercise at 31°C induced large reductions in performance (d = 0.82-1.29) and marked changes in thermoregulation, as evidenced by higher TCORE and TSKIN at fatigue, compared to exercise at 24°C. Interestingly, none of the effects induced by RSD were exacerbated by environmental heat stress and vice-versa, indicating that both conditions did not interact. We conclude that RSD and heat stress modulate aerobic performance and thermoregulatory responses by acting independently.
Collapse
Affiliation(s)
- Felipe Lioe Teh Shang
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Samuel Penna Wanner
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - William Coutinho Damasceno
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ygor Antônio Tinoco Martins
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andressa Silva
- Psychobiology and Exercise Studies Centre, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciano Sales Prado
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
22
|
Lelkes Z. Ouabain, a Na–K-ATPase inhibitor, enhances wakefulness in rats. Neuropharmacology 2020; 176:108224. [DOI: 10.1016/j.neuropharm.2020.108224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 10/23/2022]
|
23
|
Mehta R, Giri S, Mallick BN. REM sleep loss-induced elevated noradrenaline could predispose an individual to psychosomatic disorders: a review focused on proposal for prediction, prevention, and personalized treatment. EPMA J 2020; 11:529-549. [PMID: 33240449 DOI: 10.1007/s13167-020-00222-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
Historically and traditionally, it is known that sleep helps in maintaining healthy living. Its duration varies not only among individuals but also in the same individual depending on circumstances, suggesting it is a dynamic and personalized physiological process. It has been divided into rapid eye movement sleep (REMS) and non-REMS (NREMS). The former is unique that adult humans spend the least time in this stage, when although one is physically asleep, the brain behaves as if awake, the dream state. As NREMS is a pre-requisite for appearance of REMS, the latter can be considered a predictive readout of sleep quality and health. It plays a protective role against oxidative, stressful, and psychopathological insults. Several modern lifestyle activities compromise quality and quantity of sleep (including REMS) affecting fundamental physiological and psychopathosomatic processes in a personalized manner. REMS loss-induced elevated brain noradrenaline (NA) causes many associated symptoms, which are ameliorated by preventing NA action. Therefore, we propose that awareness about personalized sleep hygiene (including REMS) and maintaining optimum brain NA level should be of paramount significance for leading physical and mental well-being as well as healthy living. As sleep is a dynamic, multifactorial, homeostatically regulated process, for healthy living, we recommend addressing and treating sleep dysfunctions in a personalized manner by the health professionals, caregivers, family, and other supporting members in the society. We also recommend that maintaining sleep profile, optimum level of NA, and/or prevention of elevation of NA or its action in the brain must be seriously considered for ameliorating lifestyle and REMS disturbance-associated dysfunctions.
Collapse
Affiliation(s)
- Rachna Mehta
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India.,Present Address: Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Shatrunjai Giri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India
| | - Birendra N Mallick
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110 067 India
| |
Collapse
|
24
|
Association between autophagy and rapid eye movement sleep loss-associated neurodegenerative and patho-physio-behavioral changes. Sleep Med 2019; 63:29-37. [DOI: 10.1016/j.sleep.2019.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/26/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
|
25
|
Atrooz F, Salim S. Sleep deprivation, oxidative stress and inflammation. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:309-336. [PMID: 31997771 DOI: 10.1016/bs.apcsb.2019.03.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adequate sleep is essential for normal brain function, especially during early life developmental stages as postnatal brain maturation occurs during the critical period of childhood and adolescence. Therefore, sleep disturbance and/or deficit during this period can have detrimental consequences. Many epidemiological and clinical studies have linked early life sleep disturbance with occurrence of later life behavioral and cognitive impairments. Role of oxidative stress and inflammation has been implicated in sleep deprivation-related impairments. This review article presents a detailed description of the current state of the literature on the subject.
Collapse
Affiliation(s)
- Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
26
|
Singh A, Das G, Kaur M, Mallick BN. Noradrenaline Acting on Alpha1 Adrenoceptor as well as by Chelating Iron Reduces Oxidative Burden on the Brain: Implications With Rapid Eye Movement Sleep. Front Mol Neurosci 2019; 12:7. [PMID: 30837837 PMCID: PMC6389636 DOI: 10.3389/fnmol.2019.00007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022] Open
Abstract
The noradrenaline (NA) level in the brain is reduced during rapid eye movement sleep (REMS). However, upon REMS deprivation (REMSD) its level is elevated, which induces apoptosis and the degeneration of neurons in the brain. In contrast, isolated studies have reported that NA possesses an anti-oxidant property, while REMSD reduces lipid peroxidation (LP) and reactive oxygen species (ROS). We argued that an optimum level of NA is likely to play a physiologically beneficial role. To resolve the contradiction and for a better understanding of the role of NA in the brain, we estimated LP and ROS levels in synaptosomes prepared from the brains of control and REMS deprived rats with or without in vivo treatment with either α1-adrenoceptor (AR) antagonist, prazosin (PRZ) or α2-AR agonist, clonidine (CLN). REMSD significantly reduced LP and ROS in synaptosomes; while the effect on LP was ameliorated by both PRZ and CLN; ROS was prevented by CLN only. Thereafter, we evaluated in vitro the effects of NA, vitamin E (Vit E), vitamin C (Vit C), and desferrioxamine (DFX, iron chelator) in modulating hydrogen peroxide (H2O2)-induced LP and ROS in rat brain synaptosomes, Neuro2a, and C6 cells. We observed that NA prevented ROS generation by chelating iron (inhibiting a Fenton reaction). Also, interestingly, a lower dose of NA protected the neurons and glia, while a higher dose damaged the neurons and glia. These in vitro and in vivo results are complementary and support our contention. Based on the findings, we propose that REMS maintains an optimum level of NA in the brain (an antioxidant compromised organ) to protect the latter from continuous oxidative onslaught.
Collapse
Affiliation(s)
- Abhishek Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Gitanjali Das
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Manjeet Kaur
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
27
|
Mechanism of noradrenaline-induced α1-adrenoceptor mediated regulation of Na-K ATPase subunit expression in Neuro-2a cells. Brain Res Bull 2018; 139:157-166. [DOI: 10.1016/j.brainresbull.2018.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 02/08/2018] [Accepted: 02/20/2018] [Indexed: 01/15/2023]
|
28
|
Kuula L, Merikanto I, Makkonen T, Halonen R, Lahti-Pulkkinen M, Lahti J, Heinonen K, Räikkönen K, Pesonen AK. Schizotypal traits are associated with sleep spindles and rapid eye movement in adolescence. J Sleep Res 2018; 28:e12692. [PMID: 29655216 DOI: 10.1111/jsr.12692] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/12/2017] [Accepted: 02/27/2018] [Indexed: 01/10/2023]
Abstract
Research suggests an association between schizophrenia and a decrease in sleep spindle activity, as well as a change in sleep architecture. It is unknown how the continuum of psychotic symptoms relates to different features in the sleep electroencephalogram. We set out to examine how sleep architecture and stage 2 spindle activity are associated with schizotypy in a healthy adolescent population. The participants in our study (n = 176, 61% girls) came from a community-based cohort. Schizotypal traits were evaluated using the Schizotypal Personality Scale (STA) in early adolescence (mean age 12.3 years, SD = 0.5) and the participants underwent ambulatory overnight polysomnography at mean age 16.9 years (SD = 0.1). Sleep was scored in 30-s epochs into stages 1, 2, 3 and rapid eye movement (REM) sleep. Stage 2 spindles were detected using an automated algorithm. Spindle analyses from central and frontal derivations included spindle duration and density for slow (10-13 Hz) and fast (13-16 Hz) ranges. Covariates included sex and age. Those with the highest STA scores had a higher percentage of REM (B = 2.07 [95% CI, 0.17, 4.0]; p = .03) than those with the lowest scores. Those with the highest scores had shorter spindle duration, as derived from the frontal regions, and a slower oscillation range (B = -0.04 [95% CI, -0.07, -0.01]; p = .023) than those with the lowest scores. We conclude that high levels of schizotypy characteristics measured in early adolescence may be associated with distinguished features of sleep architecture, namely with spindle morphology and a higher proportion of REM sleep.
Collapse
Affiliation(s)
- Liisa Kuula
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilona Merikanto
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tommi Makkonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Halonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Helsinki Collegium for Advanced Studies, Helsinki, Finland
| | - Kati Heinonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu-Katriina Pesonen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Goldstein-Piekarski AN, Greer SM, Saletin JM, Harvey AG, Williams LM, Walker MP. Sex, Sleep Deprivation, and the Anxious Brain. J Cogn Neurosci 2018; 30:565-578. [PMID: 29244642 PMCID: PMC6143348 DOI: 10.1162/jocn_a_01225] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Insufficient sleep is a known trigger of anxiety. Nevertheless, not everyone experiences these effects to the same extent. One determining factor is sex, wherein women experience a greater anxiogenic impact in response to sleep loss than men. However, the underlying brain mechanism(s) governing this sleep-loss-induced anxiety increase, including the markedly different reaction in women and men, is unclear. Here, we tested the hypothesis that structural brain morphology in a discrete network of emotion-relevant regions represents one such explanatory factor. Healthy participants were assessed across sleep-rested and sleep-deprived conditions, with brain structure quantified using gray matter volume measures. Sleep loss triggered greater levels of anxiety in women compared with men. Reduced gray matter volume in the anterior insula and lateral orbitofrontal cortex predicted the anxiogenic impact of sleep loss in women, yet predicted resilience in men, and did so with high discrimination accuracy. In contrast, gray matter volume in ventromedial prefrontal cortex predicted the anxiogenic impact of sleep loss in both men and women. Structural human brain morphology therefore appears to represent one mechanistic pathway (and possible biomarker) determining anxiety vulnerability to sleep loss-a discovery that may help explain the higher prevalence of sleep disruption and anxiety in women.
Collapse
Affiliation(s)
- Andrea N. Goldstein-Piekarski
- University of California, Berkeley
- Stanford University
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC) Veterans Affairs Palo Alto Health Care System
| | | | - Jared M. Saletin
- University of California, Berkeley
- Alpert Medical School of Brown University
| | | | - Leanne M. Williams
- Stanford University
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC) Veterans Affairs Palo Alto Health Care System
| | | |
Collapse
|
30
|
Abstract
Rapid eye movement sleep (REMS) is a unique phenomenon essential for maintaining normal physiological processes and is expressed at least in species higher in the evolution. The basic scaffold of the neuronal network responsible for REMS regulation is present in the brainstem, which may be directly or indirectly influenced by most other physiological processes. It is regulated by the neurons in the brainstem. Various manipulations including chemical, elec-trophysiological, lesion, stimulation, behavioral, ontogenic and deprivation studies have been designed to understand REMS genesis, maintenance, physiology and functional significance. Although each of these methods has its significance and limitations, deprivation studies have contributed significantly to the overall understanding of REMS. In this review, we discuss the advantages and limitations of various methods used for REMS deprivation (REMSD) to understand neural regulation and physiological significance of REMS. Among the deprivation strategies, the flowerpot method is by far the method of choice because it is simple and convenient, exploits physiological parameter (muscle atonia) for REMSD and allows conducting adequate controls to overcome experimental limitations as well as to rule out nonspecific effects. Notwithstanding, a major criticism that the flowerpot method faces is that of perceived stress experienced by the experimental animals. Nevertheless, we conclude that like most methods, particularly for in vivo behavioral studies, in spite of a few limitations, given the advantages described above, the flowerpot method is the best method of choice for REMSD studies.
Collapse
Affiliation(s)
- Rachna Mehta
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Shafa Khan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
31
|
Reciprocal changes in noradrenaline and GABA levels in discrete brain regions upon rapid eye movement sleep deprivation in rats. Neurochem Int 2017; 108:190-198. [DOI: 10.1016/j.neuint.2017.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/19/2017] [Accepted: 03/24/2017] [Indexed: 12/11/2022]
|
32
|
Daniele TMDC, de Bruin PFC, Rios ERV, de Bruin VMS. Effects of exercise on depressive behavior and striatal levels of norepinephrine, serotonin and their metabolites in sleep-deprived mice. Behav Brain Res 2017; 332:16-22. [DOI: 10.1016/j.bbr.2017.05.062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/21/2017] [Accepted: 05/25/2017] [Indexed: 12/16/2022]
|
33
|
Abstract
How does a lack of sleep affect our brains? In contrast to the benefits of sleep, frameworks exploring the impact of sleep loss are relatively lacking. Importantly, the effects of sleep deprivation (SD) do not simply reflect the absence of sleep and the benefits attributed to it; rather, they reflect the consequences of several additional factors, including extended wakefulness. With a focus on neuroimaging studies, we review the consequences of SD on attention and working memory, positive and negative emotion, and hippocampal learning. We explore how this evidence informs our mechanistic understanding of the known changes in cognition and emotion associated with SD, and the insights it provides regarding clinical conditions associated with sleep disruption.
Collapse
|
34
|
Amar M, Singh A, Mallick BN. Noradrenergic β-Adrenoceptor-Mediated Intracellular Molecular Mechanism of Na-K ATPase Subunit Expression in C6 Cells. Cell Mol Neurobiol 2017; 38:441-457. [PMID: 28353187 DOI: 10.1007/s10571-017-0488-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/23/2017] [Indexed: 11/26/2022]
Abstract
Rapid eye movement sleep deprivation-associated elevated noradrenaline increases and decreases neuronal and glial Na-K ATPase activity, respectively. In this study, using C6 cell-line as a model, we investigated the possible intracellular molecular mechanism of noradrenaline-induced decreased glial Na-K ATPase activity. The cells were treated with noradrenaline in the presence or absence of adrenoceptor antagonists, modulators of extra- and intracellular Ca++ and modulators of intracellular signalling pathways. We observed that noradrenaline acting on β-adrenoceptor decreased Na-K ATPase activity and mRNA expression of the catalytic α2-Na-K ATPase subunit in the C6 cells. Further, cAMP and protein kinase-A mediated release of intracellular Ca++ played a critical role in such decreased α2-Na-K ATPase expression. In contrast, noradrenaline acting on β-adrenoceptor up-regulated the expression of regulatory β2-Na-K ATPase subunit, which although was cAMP and Ca++ dependent, was independent of protein kinase-A and protein kinase-C. Combining these with previous findings (including ours) we have proposed a working model for noradrenaline-induced suppression of glial Na-K ATPase activity and alteration in its subunit expression. The findings help understanding noradrenaline-associated maintenance of brain excitability during health and altered states, particularly in relation to rapid eye movement sleep and its deprivation when the noradrenaline level is naturally altered.
Collapse
Affiliation(s)
- Megha Amar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abhishek Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | | |
Collapse
|
35
|
Mehta R, Singh A, Mallick BN. Disciplined sleep for healthy living: Role of noradrenaline. World J Neurol 2017; 7:6-23. [DOI: 10.5316/wjn.v7.i1.6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/10/2016] [Accepted: 11/29/2016] [Indexed: 02/06/2023] Open
Abstract
Sleep is essential for maintaining normal physiological processes. It has been broadly divided into rapid eye movement sleep (REMS) and non-REMS (NREMS); one spends the least amount of time in REMS. Sleep (both NREMS and REMS) disturbance is associated with most altered states, disorders and pathological conditions. It is affected by factors within the body as well as the environment, which ultimately modulate lifestyle. Noradrenaline (NA) is one of the key molecules whose level increases upon sleep-loss, REMS-loss in particular and it induces several REMS-loss associated effects and symptoms. The locus coeruleus (LC)-NAergic neurons are primarily responsible for providing NA throughout the brain. As those neurons project to and receive inputs from across the brain, they are modulated by lifestyle changes, which include changes within the body as well as in the environment. We have reviewed the literature showing how various inputs from outside and within the body integrate at the LC neuronal level to modulate sleep (NREMS and REMS) and vice versa. We propose that these changes modulate NA levels in the brain, which in turn is responsible for acute as well as chronic psycho-somatic disorders and pathological conditions.
Collapse
|
36
|
Narwade SC, Mallick BN, Deobagkar DD. Transcriptome Analysis Reveals Altered Expression of Memory and Neurotransmission Associated Genes in the REM Sleep Deprived Rat Brain. Front Mol Neurosci 2017; 10:67. [PMID: 28367113 PMCID: PMC5355427 DOI: 10.3389/fnmol.2017.00067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Sleep disorders are associated with cognitive impairment. Selective rapid eye movement sleep (REMS) deprivation (REMSD) alters several physiological processes and behaviors. By employing NGS platform we carried out transcriptomic analysis in brain samples of control rats and those exposed to REMSD. The expression of genes involved in chromatin assembly, methylation, learning, memory, regulation of synaptic transmission, neuronal plasticity and neurohypophysial hormone synthesis were altered. Increased transcription of BMP4, DBH and ATP1B2 genes after REMSD supports our earlier findings and hypothesis. Alteration in the transcripts encoding histone subtypes and important players in chromatin remodeling was observed. The mRNAs which transcribe neurotransmitters such as OXT, AVP, PMCH and LNPEP and two small non-coding RNAs, namely RMRP and BC1 were down regulated. At least some of these changes are likely to regulate REMS and may participate in the consequences of REMS loss. Thus, the findings of this study have identified key epigenetic regulators and neuronal plasticity genes associated to REMS and its loss. This analysis provides a background and opens up avenues for unraveling their specific roles in the complex behavioral network particularly in relation to sustained REMS-loss associated changes.
Collapse
Affiliation(s)
- Santosh C Narwade
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune University Pune, India
| | | | - Deepti D Deobagkar
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune UniversityPune, India; Bioinformatics Center, Savitribai Phule Pune UniversityPune, India
| |
Collapse
|
37
|
Lee K, Baron K, Soca R, Attarian H. The Prevalence and Characteristics of REM Sleep without Atonia (RSWA) in Patients Taking Antidepressants. J Clin Sleep Med 2017; 12:351-5. [PMID: 26446247 DOI: 10.5664/jcsm.5582] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/13/2015] [Indexed: 11/13/2022]
Abstract
STUDY OBJECTIVES The association of REM sleep without atonia (RSWA) as well as REM sleep behavior disorder (RBD) with the intake of selective serotonin reuptake inhibitors (SSRI) and selective norepinephrine reuptake inhibitors (SNRI) is well established. Our study objective was to determine the prevalence of RSWA and RBD among a group of sleep center patients taking SSRI and SNRI. METHODS A retrospective chart review was done at our tertiary sleep center, and 10,746 consecutive records from October 1, 2007, through October 31, 2013, were searched for SSRI and SNRI names using the Sleep Cataloguer Software. RESULTS The search resulted in 1,444 records, which were then reviewed for keywords of RSWA and RBD. The AASM scoring criteria were used to determine RSWA. Reports of 41 patients with known narcolepsy or α-synucleinopathies were excluded. The remaining records were mined for age, sex, presence of obstructive sleep apnea (OSA), type of antidepressant (SSRI or SNRI), and diagnosis for which antidepressant was prescribed. We used logistic regression analysis to adjust for age, OSA, and sex. Of the 1,444 participants on antidepressants, 176 (12.2%) had RSWA (all confirmed by the investigators) compared to 226 of the entire sleep lab population of 10,746 (2.1%), risk ratio (95% CI) 9.978 (8.149, 12.22). Seven of the 176 patients on antidepressants had RBD (0.48%) compared to 108 of 10,746 (1%), p = 0.005. CONCLUSIONS SSRI and SNRI are associated with a higher prevalence of RSWA but not of RBD. This is independent of medication type.
Collapse
Affiliation(s)
- Kenneth Lee
- Northwestern University Feinberg School of Medicine, Chicago IL
| | - Kelly Baron
- Northwestern University Feinberg School of Medicine, Chicago IL
| | - Rodolfo Soca
- Northwestern University Feinberg School of Medicine, Chicago IL
| | - Hrayr Attarian
- Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
38
|
Khanday MA, Somarajan BI, Mehta R, Mallick BN. Noradrenaline from Locus Coeruleus Neurons Acts on Pedunculo-Pontine Neurons to Prevent REM Sleep and Induces Its Loss-Associated Effects in Rats. eNeuro 2016; 3:ENEURO.0108-16.2016. [PMID: 27957531 PMCID: PMC5144555 DOI: 10.1523/eneuro.0108-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 10/02/2016] [Accepted: 10/04/2016] [Indexed: 11/21/2022] Open
Abstract
Normally, rapid eye movement sleep (REMS) does not appear during waking or non-REMS. Isolated, independent studies showed that elevated noradrenaline (NA) levels inhibit REMS and induce REMS loss-associated cytomolecular, cytomorphological, psychosomatic changes and associated symptoms. However, the source of NA and its target in the brain for REMS regulation and function in health and diseases remained to be confirmed in vivo. Using tyrosine hydroxylase (TH)-siRNA and virus-coated TH-shRNA in normal freely moving rats, we downregulated NA synthesis in locus coeruleus (LC) REM-OFF neurons in vivo. These TH-downregulated rats showed increased REMS, which was prevented by infusing NA into the pedunculo-pontine tegmentum (PPT), the site of REM-ON neurons, normal REMS returned after recovery. Moreover, unlike normal or control-siRNA- or shRNA-injected rats, upon REMS deprivation (REMSD) TH-downregulated rat brains did not show elevated Na-K ATPase (molecular changes) expression and activity. To the best of our knowledge, these are the first in vivo findings in an animal model confirming that NA from the LC REM-OFF neurons (1) acts on the PPT REM-ON neurons to prevent appearance of REMS, and (2) are responsible for inducing REMSD-associated molecular changes and symptoms. These observations clearly show neuro-physio-chemical mechanism of why normally REMS does not appear during waking. Also, that LC neurons are the primary source of NA, which in turn causes some, if not many, REMSD-associated symptoms and behavioral changes. The findings are proof-of-principle for the first time and hold potential to be exploited for confirmation toward treating REMS disorder and amelioration of REMS loss-associated symptoms in patients.
Collapse
Affiliation(s)
| | - Bindu I Somarajan
- School of Life Sciences, Jawaharlal Nehru University , New Delhi 110607, India
| | - Rachna Mehta
- School of Life Sciences, Jawaharlal Nehru University , New Delhi 110607, India
| | | |
Collapse
|
39
|
Mehta R, Singh A, Bókkon I, Nath Mallick B. REM sleep and its Loss-Associated Epigenetic Regulation with Reference to Noradrenaline in Particular. Curr Neuropharmacol 2016; 14:28-40. [PMID: 26813120 PMCID: PMC4787282 DOI: 10.2174/1570159x13666150414185737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/02/2015] [Accepted: 04/11/2015] [Indexed: 01/12/2023] Open
Abstract
Sleep is an essential physiological process, which has been divided into rapid eye movement sleep (REMS) and non-REMS (NREMS) in higher animals. REMS is a unique phenomenon that unlike other sleep-waking states is not under voluntary control. Directly or indirectly it influences or gets influenced by most of the physiological processes controlled by the brain. It has been proposed that REMS serves housekeeping function of the brain. Extensive research has shown that during REMS at least noradrenaline (NA) -ergic neurons must cease activity and upon REMS loss, there are increased levels of NA in the brain, which then induces many of the REMS loss associated acute and chronic effects. The NA level is controlled by many bio-molecules that are regulated at the molecular and transcriptional levels. Similarly, NA can also directly or indirectly modulate the synthesis and levels of many molecules, which in turn may affect physiological processes. The burgeoning field of behavioral neuroepigenetics has gained importance in recent years and explains the regulatory mechanisms underlying several behavioral phenomena. As REMS and its loss associated changes in NA modulate several pathophysiological processes, in this review we have attempted to explain on one hand how the epigenetic mechanisms regulating the gene expression of factors like tyrosine hydroxylase (TH), monoamine oxidase (MAO), noradrenaline transporter (NAT) control NA levels and on the other hand, how NA per se can affect other molecules in neural circuitry at the epigenetic level resulting in behavioral changes in health and diseases. An
understanding of these events will expose the molecular basis of REMS and its loss-associated pathophysiological changes; which are presented as a testable hypothesis for confirmation.
Collapse
|
40
|
Association between striatal dopamine D2/D3 receptors and brain activation during visual attention: effects of sleep deprivation. Transl Psychiatry 2016; 6:e828. [PMID: 27244237 PMCID: PMC5070053 DOI: 10.1038/tp.2016.93] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 03/01/2016] [Accepted: 03/05/2016] [Indexed: 01/09/2023] Open
Abstract
Sleep deprivation (SD) disrupts dopamine (DA) signaling and impairs attention. However, the interpretation of these concomitant effects requires a better understanding of dopamine's role in attention processing. Here we test the hypotheses that D2/D3 receptors (D2/D3R) in dorsal and ventral striatum would distinctly regulate the activation of attention regions and that, by decreasing D2/D3, SD would disrupt these associations. We measured striatal D2/D3R using positron emission tomography with [(11)C]raclopride and brain activation to a visual attention (VA) task using 4-Tesla functional magnetic resonance imaging. Fourteen healthy men were studied during rested wakefulness and also during SD. Increased D2/D3R in striatum (caudate, putamen and ventral striatum) were linearly associated with higher thalamic activation. Subjects with higher D2/D3R in caudate relative to ventral striatum had higher activation in superior parietal cortex and ventral precuneus, and those with higher D2/D3R in putamen relative to ventral striatum had higher activation in anterior cingulate. SD impaired the association between striatal D2/D3R and VA-induced thalamic activation, which is essential for alertness. Findings suggest a robust DAergic modulation of cortical activation during the VA task, such that D2/D3R in dorsal striatum counterbalanced the stimulatory influence of D2/D3R in ventral striatum, which was not significantly disrupted by SD. In contrast, SD disrupted thalamic activation, which did not show counterbalanced DAergic modulation but a positive association with D2/D3R in both dorsal and ventral striatum. The counterbalanced dorsal versus ventral striatal DAergic modulation of VA activation mirrors similar findings during sensorimotor processing (Tomasi et al., 2015) suggesting a bidirectional influence in signaling between the dorsal caudate and putamen and the ventral striatum.
Collapse
|
41
|
Somarajan BI, Khanday MA, Mallick BN. Rapid Eye Movement Sleep Deprivation Induces Neuronal Apoptosis by Noradrenaline Acting on Alpha1 Adrenoceptor and by Triggering Mitochondrial Intrinsic Pathway. Front Neurol 2016; 7:25. [PMID: 27014180 PMCID: PMC4779900 DOI: 10.3389/fneur.2016.00025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/19/2016] [Indexed: 12/13/2022] Open
Abstract
Many neurodegenerative disorders are associated with rapid eye movement sleep (REMS) loss; however, the mechanism was unknown. As REMS loss elevates noradrenaline (NA) level in the brain as well as induces neuronal apoptosis and degeneration, in this study, we have delineated the intracellular molecular pathway involved in REMS deprivation (REMSD)-associated NA-induced neuronal apoptosis. Rats were REMS deprived for 6 days by the classical flower pot method; suitable controls were conducted and the effects on apoptosis markers evaluated. Further, the role of NA was studied by one, intraperitoneal (i.p.) injection of NA-ergic alpha1 adrenoceptor antagonist prazosin (PRZ) and two, by downregulation of NA synthesis in locus coeruleus (LC) neurons by local microinjection of tyrosine hydroxylase siRNA (TH-siRNA). Immunoblot estimates showed that the expressions of proapoptotic proteins viz. Bcl2-associated death promoter protein, apoptotic protease activating factor-1 (Apaf-1), cytochrome c, caspase9, caspase3 were elevated in the REMS-deprived rat brains, while caspase8 level remained unaffected; PRZ treatment did not allow elevation of these proapoptotic factors. Further, REMSD increased cytochrome c expression, which was prevented if the NA synthesis from the LC neurons was blocked by microinjection of TH-siRNA in vivo into the LC during REMSD in freely moving normal rats. Mitochondrial damage was re-confirmed by transmission electron microscopy, which showed distinctly swollen mitochondria with disintegrated cristae, chromosomal condensation, and clumping along the nuclear membrane, and all these changes were prevented in PRZ-treated rats. Combining findings of this study along with earlier reports, we propose that upon REMSD NA level increases in the brain as the LC, NA-ergic REM-OFF neurons do not cease firing and TH is upregulated in those neurons. This elevated NA acting on alpha1 adrenoceptors damages mitochondria causing release of cytochrome c to activate intrinsic pathway for inducing neuronal apoptosis in REMS-deprived rat brain.
Collapse
Affiliation(s)
- Bindu I Somarajan
- School of Life Sciences, Jawaharlal Nehru University , New Delhi , India
| | - Mudasir A Khanday
- School of Life Sciences, Jawaharlal Nehru University , New Delhi , India
| | - Birendra N Mallick
- School of Life Sciences, Jawaharlal Nehru University , New Delhi , India
| |
Collapse
|
42
|
Kitajima S, Iwata Y, Furuichi K, Sagara A, Shinozaki Y, Toyama T, Sakai N, Shimizu M, Sakurai T, Kaneko S, Wada T. Messenger RNA expression profile of sleep-related genes in peripheral blood cells in patients with chronic kidney disease. Clin Exp Nephrol 2015; 20:218-25. [DOI: 10.1007/s10157-015-1150-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 07/23/2015] [Indexed: 01/26/2023]
|
43
|
Rapid Eye Movement Sleep Deprivation Associated Increase in Na-K ATPase Activity in the Rat Brain is Due to Noradrenaline Induced α1-Adrenoceptor Mediated Increased α-Subunit of the Enzyme. Neurochem Res 2015; 40:1747-57. [DOI: 10.1007/s11064-015-1660-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
|
44
|
Gozal D, Farré R, Nieto FJ. Obstructive sleep apnea and cancer: Epidemiologic links and theoretical biological constructs. Sleep Med Rev 2015; 27:43-55. [PMID: 26447849 DOI: 10.1016/j.smrv.2015.05.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
Sleep disorders have emerged as highly prevalent conditions in the last 50-75 y. Along with improved understanding of such disorders, the realization that perturbations in sleep architecture and continuity may initiate, exacerbate or modulate the phenotypic expression of multiple diseases including cancer has gained increased attention. Furthermore, the intermittent hypoxia that is attendant to sleep disordered breathing, has recently been implicated in increased incidence and more adverse prognosis of cancer. The unifying conceptual framework linking these associations proposes that increased sympathetic activity and/or alterations in immune function, particularly affecting innate immune cellular populations, underlie the deleterious effects of sleep disorders on tumor biology. In this review, the epidemiological evidence linking disrupted sleep and intermittent hypoxia to oncological outcomes, and the potential biological underpinnings of such associations as illustrated by experimental murine models will be critically appraised. The overarching conclusion appears supportive in the formulation of an hypothetical framework, in which fragmented sleep and intermittent hypoxia may promote changes in multiple signalosomes and transcription factors that can not only initiate malignant transformation, but will also alter the tumor microenvironment, disrupt immunosurveillance, and thus hasten tumor proliferation and increase local and metastatic invasion. Future bench-based experimental studies as well as carefully conducted and controlled clinical epidemiological studies appear justified for further exploration of these hypotheses.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - F Javier Nieto
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
45
|
Effects of selective REM sleep deprivation on prefrontal gamma activity and executive functions. Int J Psychophysiol 2015; 96:115-24. [DOI: 10.1016/j.ijpsycho.2015.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/08/2015] [Accepted: 02/24/2015] [Indexed: 11/23/2022]
|
46
|
Singh A, Mallick BN. Targeting modulation of noradrenalin release in the brain for amelioration of REMS loss-associated effects. J Transl Int Med 2015; 3:8-16. [PMID: 27847879 PMCID: PMC4936468 DOI: 10.4103/2224-4018.154288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rapid eye movement sleep (REMS) loss affects most of the physiological processes, and it has been proposed that REMS maintains normal physiological processes. Changes in cultural, social, personal traits and life-style severely affect the amount and pattern of sleep, including REMS, which then manifests symptoms in animals, including humans. The effects may vary from simple fatigue and irritability to severe patho-physiological and behavioral deficits such as cognitive and behavioral dysfunctions. It has been a challenge to identify a molecule(s) that may have a potential for treating REMS loss-associated symptoms, which are very diverse. For decades, the critical role of locus coeruleus neurons in regulating REMS has been known, which has further been supported by the fact that the noradrenalin (NA) level is elevated in the brain after REMS loss. In this review, we have collected evidence from the published literature, including those from this laboratory, and argue that factors that affect REMS and vice versa modulate the level of a common molecule, the NA. Further, NA is known to affect the physiological processes affected by REMS loss. Therefore, we propose that modulation of the level of NA in the brain may be targeted for treating REMS loss-related symptoms. Further, we also argue that among the various ways to affect the release of NA-level, targeting α2 adrenoceptor autoreceptor on the pre-synaptic terminal may be the better option for ameliorating REMS loss-associated symptoms.
Collapse
Affiliation(s)
- Abhishek Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
47
|
Mehta R, Khanday MA, Mallick BN. REM sleep loss associated changes in orexin-A levels in discrete brain areas in rats. Neurosci Lett 2015; 590:62-7. [PMID: 25637698 DOI: 10.1016/j.neulet.2015.01.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/27/2015] [Indexed: 10/24/2022]
Abstract
Rapid eye movement sleep (REMS) serves house-keeping function of the brain and its loss affects several pathophysiological processes. Relative levels of neurotransmitters including orexin A (Orx-A) in various parts of the brain in health and diseases are among the key factors for modulation of behaviors, including REMS. The level of neurotransmitter in an area in the brain directly depends on number of projecting neurons and their firing rates. The locus coeruleus (LC), the site of REM-OFF neurons, receives densest, while the pedunculo-pontine area (PPT), the site of REM-ON neurons receives lesser projections from the Orx-ergic neurons. Further, the Orx-ergic neurons are active during waking and silent during REMS and NREMS. Therefore, the level of Orx-A in discrete regions of the brain is likely to be different during normal and altered states, which in turn is likely to be responsible for altered behaviors in health and diseases, including in relation to REMS. Therefore, in the present study, we estimated Orx-A level in LC, cortex, posterior hypothalamus (PH), hippocampus, and PPT after 96 h REMSD, in post-deprivation recovered rats and in control rats. This is the first report of estimation of Orx-A in different brain regions after prolonged REMSD. It was observed that after REMSD the Orx-A level increased significantly in LC, cortex and PH which returned to normal level after recovery; however, the level did not change in the hippocampus and PPT. The Orx-A induced modulation of REMS could be secondary to increased waking.
Collapse
Affiliation(s)
- Rachna Mehta
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | | | |
Collapse
|
48
|
Elliott AS, Huber JD, O'Callaghan JP, Rosen CL, Miller DB. A review of sleep deprivation studies evaluating the brain transcriptome. SPRINGERPLUS 2014; 3:728. [PMID: 25932362 PMCID: PMC4409616 DOI: 10.1186/2193-1801-3-728] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/25/2014] [Indexed: 12/14/2022]
Abstract
Epidemiological studies show a positive association between adequate sleep and good health. Further, disrupted sleep may increase the risk for CNS diseases, such as stroke and Alzheimer’s disease. However, there has been limited progress in determining how sleep is linked to brain health or how sleep disruption may increase susceptibility to brain insult and disease. Animal studies can aid in understanding these links. In reviewing the animal literature related to the effects of sleep disruption on the brain, we found most of the work was directed toward investigating and characterizing the role of various brain areas or structures in initiating and regulating sleep. In contrast, limited effort has been directed towards understanding how sleep disruption alters the brain’s health or susceptibility to insult. We also note many current studies have determined the changes in the brain following compromised sleep by examining, for example, the brain transcriptome or to a more limited extent the proteome. However, these studies have utilized almost exclusively total sleep deprivation (e.g., 24 out of 24 hours) paradigms or single short periods of limited acute sleep deprivation (e.g., 3 out of 24 hours). While such strategies are beneficial in understanding how sleep is controlled, they may not have much translational value for determining links between sleep and brain health or for determining how sleep disruption may increase brain susceptibility to insult. Surprisingly, few studies have determined how the duration and recurrence of sleep deprivation influence the effects seen after sleep deprivation. Our aim in this review was to identify relevant rodent studies from 1980 through 2012 and analyze those that use varying durations of sleep deprivation or restriction in their effort to evaluate the effects of sleep deprivation on the brain transcriptome and to a more limited extent the proteome. We examined how differences in the duration of sleep deprivation affect gene and protein expression to better understand the full consequences of repeated sleep disruption on the brain. Future research needs to consider and emphasize how the type and extent of the sleep deprivation exposure impacts the conclusions reached concerning the influence of sleep disruption on the brain. We identified relevant studies between 1980 and 2012 by searching the electronic databases of PubMed, Medline (Ovid), Embase (Ovid), and Web of Science using the terms “sleep” AND “disrupt”, “deprivation”, “restrict”, “fragment”, “loss”, “disturb”, “disorder”, “dysfunction”, “brain”, “cortex”, striatum”, hypothalamus”, “hippocampus”, “gene”, “protein”, “genomics”, “proteomics”, “polymerase chain reaction”, “pcr”, “microarray”, “molecular”, “rodent” “rat”, “rats”, “mouse”, “mice”. All searches were limited to rodent studies in English and the reference lists of retrieved articles were searched for additional pertinent studies.
Collapse
Affiliation(s)
- Alisa S Elliott
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Jason D Huber
- School of Pharmacy, West Virginia University, Morgantown, WV USA
| | - James P O'Callaghan
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| | - Charles L Rosen
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Diane B Miller
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| |
Collapse
|
49
|
Jaseja H. Deep-brain stimulation in intractable epilepsy: pedunculopontine nucleus versus thalamic nuclei: a perspective. World Neurosurg 2014; 82:e568-9. [PMID: 24834872 DOI: 10.1016/j.wneu.2014.05.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 05/09/2014] [Indexed: 02/05/2023]
|
50
|
Peters AC, Blechert J, Sämann PG, Eidner I, Czisch M, Spoormaker VI. One night of partial sleep deprivation affects habituation of hypothalamus and skin conductance responses. J Neurophysiol 2014; 112:1267-76. [PMID: 24920020 DOI: 10.1152/jn.00657.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sleep disturbances are prevalent in clinical anxiety, but it remains unclear whether they are cause and/or consequence of this condition. Fear conditioning constitutes a valid laboratory model for the acquisition of normal and pathological anxiety. To explore the relationship between disturbed sleep and anxiety in more detail, the present study evaluated the effect of partial sleep deprivation (SD) on fear conditioning in healthy individuals. The neural correlates of 1) nonassociative learning and physiological processing and 2) associative learning (differential fear conditioning) were addressed. Measurements entailed simultaneous functional MRI, EEG, skin conductance response (SCR), and pulse recordings. Regarding nonassociative learning, partial SD resulted in a generalized failure to habituate during fear conditioning, as evidenced by reduced habituation of SCR and hypothalamus responses to all stimuli. Furthermore, SCR and hypothalamus activity were correlated, supporting their functional relationship. Regarding associative learning, effects of partial SD on the acquisition of conditioned fear were weaker and did not reach statistical significance. The hypothalamus plays an integral role in the regulation of sleep and autonomic arousal. Thus sleep disturbances may play a causal role in the development of normal and possibly pathological fear by increasing the susceptibility of the sympathetic nervous system to stressful experiences.
Collapse
Affiliation(s)
- Anja C Peters
- Neuroimaging Research Group, Max Planck Institute of Psychiatry, Munich, Germany; and
| | - Jens Blechert
- Division of Clinical Psychology, Psychotherapy, and Health Psychology, Institute of Psychology, University of Salzburg, Salzburg, Austria
| | - Philipp G Sämann
- Neuroimaging Research Group, Max Planck Institute of Psychiatry, Munich, Germany; and
| | - Ines Eidner
- Neuroimaging Research Group, Max Planck Institute of Psychiatry, Munich, Germany; and
| | - Michael Czisch
- Neuroimaging Research Group, Max Planck Institute of Psychiatry, Munich, Germany; and
| | - Victor I Spoormaker
- Neuroimaging Research Group, Max Planck Institute of Psychiatry, Munich, Germany; and
| |
Collapse
|