1
|
Yue Y, Zhang X, Lv W, Lai HY, Shen T. Interplay between the glymphatic system and neurotoxic proteins in Parkinson's disease and related disorders: current knowledge and future directions. Neural Regen Res 2024; 19:1973-1980. [PMID: 38227524 DOI: 10.4103/1673-5374.390970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
Parkinson's disease is a common neurodegenerative disorder that is associated with abnormal aggregation and accumulation of neurotoxic proteins, including α-synuclein, amyloid-β, and tau, in addition to the impaired elimination of these neurotoxic protein. Atypical parkinsonism, which has the same clinical presentation and neuropathology as Parkinson's disease, expands the disease landscape within the continuum of Parkinson's disease and related disorders. The glymphatic system is a waste clearance system in the brain, which is responsible for eliminating the neurotoxic proteins from the interstitial fluid. Impairment of the glymphatic system has been proposed as a significant contributor to the development and progression of neurodegenerative disease, as it exacerbates the aggregation of neurotoxic proteins and deteriorates neuronal damage. Therefore, impairment of the glymphatic system could be considered as the final common pathway to neurodegeneration. Previous evidence has provided initial insights into the potential effect of the impaired glymphatic system on Parkinson's disease and related disorders; however, many unanswered questions remain. This review aims to provide a comprehensive summary of the growing literature on the glymphatic system in Parkinson's disease and related disorders. The focus of this review is on identifying the manifestations and mechanisms of interplay between the glymphatic system and neurotoxic proteins, including loss of polarization of aquaporin-4 in astrocytic endfeet, sleep and circadian rhythms, neuroinflammation, astrogliosis, and gliosis. This review further delves into the underlying pathophysiology of the glymphatic system in Parkinson's disease and related disorders, and the potential implications of targeting the glymphatic system as a novel and promising therapeutic strategy.
Collapse
Affiliation(s)
- Yumei Yue
- Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiaodan Zhang
- Department of Emergency Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Lv
- Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hsin-Yi Lai
- Department of Neurology of the Second Affiliated Hospital and School of Brain Science and Brain Medicine, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang Province, China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ting Shen
- Department of Neurology of the Second Affiliated Hospital and School of Brain Science and Brain Medicine, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Jin RN, Inada H, Momma H, Ma D, Yuan K, Nagatomi R. Impact of carbon dioxide exposures on sleep latency among healthy volunteers: A randomized order, paired crossover study, evidence from the multiple sleep latency test. ENVIRONMENTAL RESEARCH 2024; 262:119785. [PMID: 39142454 DOI: 10.1016/j.envres.2024.119785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Daytime sleepiness affects work efficiency, occupational safety, and public health. Although previous studies have reported an association between environmental carbon dioxide (eCO2) and daytime sleepiness, it has been challenging to draw a firm conclusion due to the lack of standardized sampling and profiling protocols. OBJECTIVE We examined the effect of pure CO2 exposure at 5000 (ppm, parts per million) on daytime sleepiness. METHODS Eleven healthy participants (males of 24 ± 3 years, mean ± SD) completed a four-nap multiple sleep latency test (MSLT) protocol in the environmentally controlled chamber under two conditions: the CO2 condition (4851 ± 229 ppm) and the Control condition (1102 ± 204 ppm). The subjective sleepiness level and cognitive performances were also evaluated using the Stanford Sleepiness Scale (SSS) questionnaire, Psychomotor Vigilance Test (PVT), and Stroop test after each nap session. RESULTS A significant reduction in sleep latency was observed in the CO2 exposure condition (Control vs. CO2 = 13.1 ± 3.3 min vs. 9.7 ± 3.2 min). The subjective sleepiness scores were also significantly higher in the CO2 exposure condition than in the Control condition (Control vs. CO2 = 2.7 ± 0.5 vs. 4.7 ± 0.8). Cognitive responses after naps showed no significant difference across conditions. CONCLUSION This study revealed that exposure to environmental CO2 at a concentration as high as the upper safety limit at work sites significantly shortened the sleep latency and enhanced subjective sleepiness during naps in the MSLT without affecting cognitive responses after each exposure. Our results demonstrated that exposure to high environmental CO2 induces daytime sleepiness that potentially compromises work efficiency and safety.
Collapse
Affiliation(s)
- Rui Nian Jin
- Division of Biomedical Engineering for Health & Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Miyagi, 980-8575, Japan; Tohoku University, Designing Future Health Initiative, Promotion Office of Strategic Innovation, Sendai, Miyagi, 980-0845, Japan
| | - Hitoshi Inada
- Division of Biomedical Engineering for Health & Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Miyagi, 980-8575, Japan; Department of Biochemistry & Cellular Biology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502, Japan.
| | - Haruki Momma
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Dongmei Ma
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Keqing Yuan
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Ryoichi Nagatomi
- Division of Biomedical Engineering for Health & Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Miyagi, 980-8575, Japan; Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan; Tohoku University, Designing Future Health Initiative, Promotion Office of Strategic Innovation, Sendai, Miyagi, 980-0845, Japan.
| |
Collapse
|
3
|
Aframian K, Yousef Yengej D, Nwaobi S, Raman S, Faas GC, Charles A. Effects of chronic caffeine on patterns of brain blood flow and behavior throughout the sleep-wake cycle in freely behaving mice. PNAS NEXUS 2023; 2:pgad303. [PMID: 37780231 PMCID: PMC10538474 DOI: 10.1093/pnasnexus/pgad303] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
Caffeine has significant effects on neurovascular activity and behavior throughout the sleep-wake cycle. We used a minimally invasive microchip/video system to continuously record effects of caffeine in the drinking water of freely behaving mice. Chronic caffeine shifted both rest and active phases by up to 2 h relative to the light-dark cycle in a dose-dependent fashion. There was a particular delay in the onset of rapid eye movement (REM) sleep as compared with non-REM sleep during the rest phase. Chronic caffeine increased wakefulness during the active phase and consolidated sleep during the rest phase; overall, there was no net change in the amount of time spent in the wake, sleep, or REM sleep states during caffeine administration. Despite these effects on wakefulness and sleep, chronic caffeine decreased mean cerebral blood volume (CBV) during the active phase and increased mean CBV during the rest phase. Chronic caffeine also increased heart rate variability in both the sleep and wake states. These results provide new insight into the effects of caffeine on the biology of the sleep-wake cycle. Increased blood flow during sleep caused by chronic caffeine may have implications for its potential neuroprotective effects through vascular mechanisms of brain waste clearance.
Collapse
Affiliation(s)
- Kimiya Aframian
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Dmitri Yousef Yengej
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Sinifunanya Nwaobi
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Shrayes Raman
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Guido C Faas
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Andrew Charles
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Nollet M, Franks NP, Wisden W. Understanding Sleep Regulation in Normal and Pathological Conditions, and Why It Matters. J Huntingtons Dis 2023; 12:105-119. [PMID: 37302038 PMCID: PMC10473105 DOI: 10.3233/jhd-230564] [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] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
Sleep occupies a peculiar place in our lives and in science, being both eminently familiar and profoundly enigmatic. Historically, philosophers, scientists and artists questioned the meaning and purpose of sleep. If Shakespeare's verses from MacBeth depicting "Sleep that soothes away all our worries" and "relieves the weary laborer and heals hurt minds" perfectly epitomize the alleviating benefits of sleep, it is only during the last two decades that the growing understanding of the sophisticated sleep regulatory mechanisms allows us to glimpse putative biological functions of sleep. Sleep control brings into play various brain-wide processes occurring at the molecular, cellular, circuit, and system levels, some of them overlapping with a number of disease-signaling pathways. Pathogenic processes, including mood disorders (e.g., major depression) and neurodegenerative illnesses such Huntington's or Alzheimer's diseases, can therefore affect sleep-modulating networks which disrupt the sleep-wake architecture, whereas sleep disturbances may also trigger various brain disorders. In this review, we describe the mechanisms underlying sleep regulation and the main hypotheses drawn about its functions. Comprehending sleep physiological orchestration and functions could ultimately help deliver better treatments for people living with neurodegenerative diseases.
Collapse
Affiliation(s)
- Mathieu Nollet
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - Nicholas P. Franks
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - William Wisden
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
5
|
Verghese JP, Terry A, de Natale ER, Politis M. Research Evidence of the Role of the Glymphatic System and Its Potential Pharmacological Modulation in Neurodegenerative Diseases. J Clin Med 2022; 11:jcm11236964. [PMID: 36498538 PMCID: PMC9735716 DOI: 10.3390/jcm11236964] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
The glymphatic system is a unique pathway that utilises end-feet Aquaporin 4 (AQP4) channels within perivascular astrocytes, which is believed to cause cerebrospinal fluid (CSF) inflow into perivascular space (PVS), providing nutrients and waste disposal of the brain parenchyma. It is theorised that the bulk flow of CSF within the PVS removes waste products, soluble proteins, and products of metabolic activity, such as amyloid-β (Aβ). In the experimental model, the glymphatic system is selectively active during slow-wave sleep, and its activity is affected by both sleep dysfunction and deprivation. Dysfunction of the glymphatic system has been proposed as a potential key driver of neurodegeneration. This hypothesis is indirectly supported by the close relationship between neurodegenerative diseases and sleep alterations, frequently occurring years before the clinical diagnosis. Therefore, a detailed characterisation of the function of the glymphatic system in human physiology and disease would shed light on its early stage pathophysiology. The study of the glymphatic system is also critical to identifying means for its pharmacological modulation, which may have the potential for disease modification. This review will critically outline the primary evidence from literature about the dysfunction of the glymphatic system in neurodegeneration and discuss the rationale and current knowledge about pharmacological modulation of the glymphatic system in the animal model and its potential clinical applications in human clinical trials.
Collapse
|
6
|
Abstract
A molecular circadian clock exists not only in the brain, but also in most cells of the body. Research over the past two decades has demonstrated that it directs daily rhythmicity of nearly every aspect of metabolism. It also consolidates sleep-wake behavior each day into an activity/feeding period and a sleep/fasting period. Otherwise, sleep-wake states are mostly controlled by hypothalamic and thalamic regulatory circuits in the brain that direct overall brain state. Recent evidence suggests that hypothalamic control of appetite and metabolism may be concomitant with sleep-wake regulation, and even share the same control centers. Thus, circadian control of metabolic pathways might be overlaid by sleep-wake control of the same pathways, providing a flexible and redundant system to modify metabolism according to both activity and environment.
Collapse
|
7
|
Giovanni A, Giorgia A. The neurophysiological basis of bruxism. Heliyon 2021; 7:e07477. [PMID: 34286138 PMCID: PMC8273205 DOI: 10.1016/j.heliyon.2021.e07477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/05/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022] Open
Abstract
Mesencephalic trigeminal nucleus (MTN) neurons innervate the stretch receptors of the jaw elevator muscles and periodontal ligament mechanoreceptors, Bruxism activates the MTN. We analyzed how MTN cells are structured, their anatomy and physiology, and the effects of their activation. To induce and maintain sleep, gamma-aminobutyric acid (GABA), an inhibitor neurotransmitter, is released from the ventro-lateral preoptic area of the hypothalamus and acts on the ascending reticular activating system (ARAS) nuclei. The GABA neurotrasmitter induces the entry of chlorine into cells, hyperpolarizing and inhibiting these. MTN cells, on the contrary, are depolarized by GABA, as their receptors are activated upon GABA binding. They “let out” chlorine and activate ARAS cells. MTN cells release glutamate, an excitatory neurotransmitter onto their target cells, in this case onto ARAS cells. During wakefulness, ARAS activation causes cerebral cortex activation; instead, during sleep (sleep bruxism), ARAS activation avoids an excessive reduction in ARAS neurotransmitters, including noradrenaline, dopamine, serotonin, acetylcholine and glutamate. These neurotransmitters, in addition to activating the cerebral cortex, modulate vital functions such as cardiac and respiratory functions. Polysomnography shows that sleep bruxism is always accompanied by cardiac and respiratory activation and, most importantly, by brain function activation. Bruxism is not a parafunction, and it functions to activate ARAS nuclei.
Collapse
Affiliation(s)
- Andrisani Giovanni
- Matera, via della Croce 47, Italy.,Ezelsveldlaan 2, 2611 rv, Delft, the Netherlands
| | | |
Collapse
|
8
|
Pires GN, Oliveira TB, Mello VFF, Bezerra AG, Leenaars CHC, Ritskes-Hoitinga M, Tufik S, Andersen ML. Effects of sleep deprivation on maternal behaviour in animal models: A systematic review and meta-analysis. J Sleep Res 2021; 30:e13333. [PMID: 33719116 DOI: 10.1111/jsr.13333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 01/12/2023]
Abstract
Pregnancy is a period of numerous physical and emotional changes in women's lives, including alterations in sleep patterns and worsening of pre-existing sleep disturbances, which possibly lead to impaired postpartum maternal behaviour and mother-infant relationship. The effects of sleep deprivation during pregnancy in maternal behaviour have been evaluated in preclinical studies, but have provided inconsistent results. Thus, in the present study, we aimed to evaluate the effects of sleep deprivation during pregnancy on maternal behaviour of animals through a systematic review and meta-analyses. After a two-step selection process, six articles were included, all of them describing rat studies. The most frequently used method of sleep deprivation was rapid eye movement sleep restriction, using the multiple-platform method. Four meta-analyses were performed, none of them presenting significant impact of sleep deprivation on maternal behaviour, failing to reproduce the results observed in previous clinical studies. In conclusion, our results show a lack of translational applicability of animal models to evaluate the effects of sleep deprivation during pregnancy on maternal behaviour.
Collapse
Affiliation(s)
- Gabriel N Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Thainá B Oliveira
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Victoria F F Mello
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Andréia G Bezerra
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Merel Ritskes-Hoitinga
- Systematic Review Centre for Laboratory (animal) Experimentation (SYRCLE), Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands.,AUGUST, Department for Clinical Medicine, Aarhus University, Denmark
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Gomes JI, Farinha-Ferreira M, Rei N, Gonçalves-Ribeiro J, Ribeiro JA, Sebastião AM, Vaz SH. Of adenosine and the blues: The adenosinergic system in the pathophysiology and treatment of major depressive disorder. Pharmacol Res 2020; 163:105363. [PMID: 33285234 DOI: 10.1016/j.phrs.2020.105363] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Major depressive disorder (MDD) is the foremost cause of global disability, being responsible for enormous personal, societal, and economical costs. Importantly, existing pharmacological treatments for MDD are partially or totally ineffective in a large segment of patients. As such, the search for novel antidepressant drug targets, anchored on a clear understanding of the etiological and pathophysiological mechanisms underpinning MDD, becomes of the utmost importance. The adenosinergic system, a highly conserved neuromodulatory system, appears as a promising novel target, given both its regulatory actions over many MDD-affected systems and processes. With this goal in mind, we herein review the evidence concerning the role of adenosine as a potential player in pathophysiology and treatment of MDD, combining data from both human and animal studies. Altogether, evidence supports the assertions that the adenosinergic system is altered in both MDD patients and animal models, and that drugs targeting this system have considerable potential as putative antidepressants. Furthermore, evidence also suggests that modifications in adenosine signaling may have a key role in the effects of several pharmacological and non-pharmacological antidepressant treatments with demonstrated efficacy, such as electroconvulsive shock, sleep deprivation, and deep brain stimulation. Lastly, it becomes clear from the available literature that there is yet much to study regarding the role of the adenosinergic system in the pathophysiology and treatment of MDD, and we suggest several avenues of research that are likely to prove fruitful.
Collapse
Affiliation(s)
- Joana I Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Farinha-Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
10
|
Zhang D, Jin W, Liu H, Liang T, Peng Y, Zhang J, Zhang Y. ENT1 inhibition attenuates apoptosis by activation of cAMP/pCREB/Bcl2 pathway after MCAO in rats. Exp Neurol 2020; 331:113362. [PMID: 32445645 DOI: 10.1016/j.expneurol.2020.113362] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND PURPOSE The present study was designed to investigate the potential role and the mechanism of equilibrative nucleoside transporter 1 (ENT1) on neuronal apoptosis and neurological deficits after middle cerebral artery occlusion (MCAO) in rats. METHODS One hundred and thirty-four male Sprague-Dawley rats were subjected to two hours of MCAO followed by reperfusion. The time course of the expression level of ENT1 and phosphorylation of CREB were detected by western blot and immunofluorescence staining. Another set of animals were administrated with NBTI, the ENT1 inhibitor, by daily intraperitoneal injection starting at 0.5 h post-MCAO, infarction volume and neurological deficits were measured both at 24 h and 72 h post MCAO. We further explored the neuroprotection machenism by using H89, cAMP dependent protein kinase inhibitor, the expression of Bcl-2, Bax, phosphorylated CREB and Cleaved caspase-3 were quantified by Western blot, neuronal apoptosis were analyed by TUNEL staining. RESULTS The endogenous expression of ENT1 were significantly increased and peaked at 12 h after MCAO. High-dose of NBTI (15 mg/kg) reduced brain infarction volume and improved neurologic deficits both at 24 h and 72 h post MCAO. Moreover, NBTI significantly increased the level of CREB phosphorylation and extracellular adenosine concentration, and decreased the neuronal apoptosis 24 h after MCAO. NBTI treatment reduced the expression of Bax and cleaved caspase-3, while up-regulated Bcl-2 compared with vehicle group. These effects were abolished by H89 pretreatment. CONCLUSIONS ENT1 inhibition prevented neuronal apoptosis and improves neurological deficits through cAMP/PKA/CREB/Bcl-2 signaling pathway after MCAO in rats. ENT1 might be an effective target in the treatment strategy for ischemic stroke.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of rehabilitation medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Weidong Jin
- Department of Laboratory Medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongliang Liu
- Department of rehabilitation medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Liang
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Yan Peng
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China
| | - Jun Zhang
- Department of neurology, Affiliated Hospital, Zunyi medical University, Zunyi, China.
| | - Yang Zhang
- Department of Laboratory Medicine, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
11
|
Brain Glymphatic/Lymphatic Imaging by MRI and PET. Nucl Med Mol Imaging 2020; 54:207-223. [PMID: 33088350 DOI: 10.1007/s13139-020-00665-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/09/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
Since glymphatic was proposed and meningeal lymphatic was discovered, MRI and even PET were introduced to investigate brain parenchymal interstitial fluid (ISF), cerebrospinal fluid (CSF), and lymphatic outflow in rodents and humans. Previous findings by ex vivo fluorescent microscopic, and in vivo two-photon imaging in rodents were reproduced using intrathecal contrast (gadobutrol and the similar)-enhanced MRI in rodents and further in humans. On dynamic MRI of meningeal lymphatics, in contrast to rodents, humans use mainly dorsal meningeal lymphatic pathways of ISF-CSF-lymphatic efflux. In mice, ISF-CSF exchange was examined thoroughly using an intra-cistern injection of fluorescent tracers during sleep, aging, and neurodegeneration yielding many details. CSF to lymphatic efflux is across arachnoid barrier cells over the dorsal dura in rodents and in humans. Meningeal lymphatic efflux to cervical lymph nodes and systemic circulation is also well-delineated especially in humans onintrathecal contrast MRI. Sleep- or anesthesia-related changes of glymphatic-lymphatic flow and the coupling of ISF-CSF-lymphatic drainage are major confounders ininterpreting brain glymphatic/lymphatic outflow in rodents. PET imaging in humans should be interpreted based on human anatomy and physiology, different in some aspects, using MRI recently. Based on the summary in this review, we propose non-invasive and longer-term intrathecal SPECT/PET or MRI studies to unravel the roles of brain glymphatic/lymphatic in diseases.
Collapse
|
12
|
Negro A, Seidel JL, Houben T, Yu ES, Rosen I, Arreguin AJ, Yalcin N, Shorser-Gentile L, Pearlman L, Sadhegian H, Vetrivelan R, Chamberlin NL, Ayata C, Martelletti P, Moskowitz MA, Eikermann-Haerter K. Acute sleep deprivation enhances susceptibility to the migraine substrate cortical spreading depolarization. J Headache Pain 2020; 21:86. [PMID: 32631251 PMCID: PMC7339460 DOI: 10.1186/s10194-020-01155-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Migraine is a common headache disorder, with cortical spreading depolarization (CSD) considered as the underlying electrophysiological event. CSD is a slowly propagating wave of neuronal and glial depolarization. Sleep disorders are well known risk factors for migraine chronification, and changes in wake-sleep pattern such as sleep deprivation are common migraine triggers. The underlying mechanisms are unknown. As a step towards developing an animal model to study this, we test whether sleep deprivation, a modifiable migraine trigger, enhances CSD susceptibility in rodent models. METHODS Acute sleep deprivation was achieved using the "gentle handling method", chosen to minimize stress and avoid confounding bias. Sleep deprivation was started with onset of light (diurnal lighting conditions), and assessment of CSD was performed at the end of a 6 h or 12 h sleep deprivation period. The effect of chronic sleep deprivation on CSD was assessed 6 weeks or 12 weeks after lesioning of the hypothalamic ventrolateral preoptic nucleus. All experiments were done in a blinded fashion with respect to sleep status. During 60 min of continuous topical KCl application, we assessed the total number of CSDs, the direct current shift amplitude and duration of the first CSD, the average and cumulative duration of all CSDs, propagation speed, and electrical CSD threshold. RESULTS Acute sleep deprivation of 6 h (n = 17) or 12 h (n = 11) duration significantly increased CSD frequency compared to controls (17 ± 4 and 18 ± 2, respectively, vs. 14 ± 2 CSDs/hour in controls; p = 0.003 for both), whereas other electrophysiological properties of CSD were unchanged. Acute total sleep deprivation over 12 h but not over 6 h reduced the electrical threshold of CSD compared to controls (p = 0.037 and p = 0.095, respectively). Chronic partial sleep deprivation in contrast did not affect CSD susceptibility in rats. CONCLUSIONS Acute but not chronic sleep deprivation enhances CSD susceptibility in rodents, possibly underlying its negative impact as a migraine trigger and exacerbating factor. Our findings underscore the importance of CSD as a therapeutic target in migraine and suggest that headache management should identify and treat associated sleep disorders.
Collapse
Affiliation(s)
- Andrea Negro
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Jessica L Seidel
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Thijs Houben
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Esther S Yu
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ike Rosen
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Andrea J Arreguin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Nilufer Yalcin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Lea Shorser-Gentile
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Lea Pearlman
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Homa Sadhegian
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Michael A Moskowitz
- Department of Radiology, and Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katharina Eikermann-Haerter
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
13
|
Murillo-Rodríguez E, Arankowsky-Sandoval G, Pertwee RG, Parker L, Mechoulam R. Sleep and neurochemical modulation by cannabidiolic acid methyl ester in rats. Brain Res Bull 2020; 155:166-173. [DOI: 10.1016/j.brainresbull.2019.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/25/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023]
|
14
|
Sun Q, Shi J, Ni J, Wei M, Zhang X, Li T, Wang P, Guo B, Tian J. Efficacy of Chinese herbal compound GAPT for the treatment of Alzheimer’s disease during mouse/rat studies: A systematic review and meta-analysis. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2019. [DOI: 10.1016/j.jtcms.2019.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
15
|
Ma Y, Miracca G, Yu X, Harding EC, Miao A, Yustos R, Vyssotski AL, Franks NP, Wisden W. Galanin Neurons Unite Sleep Homeostasis and α2-Adrenergic Sedation. Curr Biol 2019; 29:3315-3322.e3. [PMID: 31543455 PMCID: PMC6868514 DOI: 10.1016/j.cub.2019.07.087] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/24/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022]
Abstract
Our urge to sleep increases with time spent awake, until sleep becomes inescapable. The sleep following sleep deprivation is longer and deeper, with an increased power of delta (0.5–4 Hz) oscillations, a phenomenon termed sleep homeostasis [1, 2, 3, 4]. Although widely expressed genes regulate sleep homeostasis [1, 4, 5, 6, 7, 8, 9, 10] and the process is tracked by somnogens and phosphorylation [1, 3, 7, 11, 12, 13, 14], at the circuit level sleep homeostasis has remained mysterious. Previously, we found that sedation induced with α2-adrenergic agonists (e.g., dexmedetomidine) and sleep homeostasis both depend on the preoptic (PO) hypothalamus [15, 16]. Dexmedetomidine, increasingly used for long-term sedation in intensive care units [17], induces a non-rapid-eye-movement (NREM)-like sleep but with undesirable hypothermia [18, 19]. Within the PO, various neuronal subtypes (e.g., GABA/galanin and glutamate/NOS1) induce NREM sleep [20, 21, 22] and concomitant body cooling [21, 22]. This could be because NREM sleep’s restorative effects depend on lower body temperature [23, 24]. Here, we show that mice with lesioned PO galanin neurons have reduced sleep homeostasis: in the recovery sleep following sleep deprivation there is a diminished increase in delta power, and the mice catch up little on lost sleep. Furthermore, dexmedetomidine cannot induce high-power delta oscillations or sustained hypothermia. Some hours after dexmedetomidine administration to wild-type mice there is a rebound in delta power when they enter normal NREM sleep, reminiscent of emergence from torpor. This delta rebound is reduced in mice lacking PO galanin neurons. Thus, sleep homeostasis and dexmedetomidine-induced sedation require PO galanin neurons and likely share common mechanisms. This is the first identification of a cell type underlying sleep homeostasis Preoptic galanin neurons are essential for sleep homeostasis Galanin neurons mediate the sedative and hypothermic actions of dexmedetomidine Dexmedetomidine causes an EEG delta power rebound dependent on galanin neurons
Collapse
Affiliation(s)
- Ying Ma
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Giulia Miracca
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Xiao Yu
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Edward C Harding
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Andawei Miao
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College, London SW7 2AZ, UK
| | - Raquel Yustos
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich 8057, Switzerland
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College, London SW7 2AZ, UK.
| | - William Wisden
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College, London SW7 2AZ, UK.
| |
Collapse
|
16
|
The Neuropeptide Galanin Is Required for Homeostatic Rebound Sleep following Increased Neuronal Activity. Neuron 2019; 104:370-384.e5. [PMID: 31537465 DOI: 10.1016/j.neuron.2019.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 06/04/2019] [Accepted: 08/03/2019] [Indexed: 01/19/2023]
Abstract
Sleep pressure increases during wake and dissipates during sleep, but the molecules and neurons that measure homeostatic sleep pressure remain poorly understood. We present a pharmacological assay in larval zebrafish that generates short-term increases in wakefulness followed by sustained rebound sleep after washout. The intensity of global neuronal activity during drug-induced wakefulness predicted the amount of subsequent rebound sleep. Whole-brain mapping with the neuronal activity marker phosphorylated extracellular signal-regulated kinase (pERK) identified preoptic Galanin (Galn)-expressing neurons as selectively active during rebound sleep, and the relative induction of galn transcripts was predictive of total rebound sleep time. Galn is required for sleep homeostasis, as galn mutants almost completely lacked rebound sleep following both pharmacologically induced neuronal activity and physical sleep deprivation. These results suggest that Galn plays a key role in responding to sleep pressure signals derived from neuronal activity and functions as an output arm of the vertebrate sleep homeostat.
Collapse
|
17
|
Calker D, Biber K, Domschke K, Serchov T. The role of adenosine receptors in mood and anxiety disorders. J Neurochem 2019; 151:11-27. [DOI: 10.1111/jnc.14841] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Dietrich Calker
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Knut Biber
- Section Medical Physiology, Department of Neuroscience University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Katharina Domschke
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
- Centre for Basics in Neuromodulation, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine, Medical Center ‐ University Freiburg University of Freiburg Freiburg Germany
| |
Collapse
|
18
|
Sleep and Microdialysis: An Experiment and a Systematic Review of Histamine and Several Amino Acids. J Circadian Rhythms 2019; 17:7. [PMID: 31303885 PMCID: PMC6611484 DOI: 10.5334/jcr.183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sleep seems essential to proper functioning of the prefrontal cortex (PFC). The role of different neurotransmitters has been studied, mainly the catecholamines and serotonin. Less attention has been paid to the amino acid transmitters and histamine. Here, we focus on the activity of these molecules in the PFC during sleep and sleep deprivation (SD). We determined extracellular concentrations of histamine and 8 amino acids in the medial PFC before, during and after SD. Additionally, we systematically reviewed the literature on studies reporting microdialysis measurements relating to sleep throughout the brain. In our experiment, median concentrations of glutamate were higher during SD than during baseline (p = 0.013) and higher during the dark-active than during the resting phase (p = 0.003). Glutamine was higher during post-SD recovery than during baseline (p = 0.010). For other compounds, no differences were observed between light and dark circadian phase, and between sleep deprivation, recovery and baseline. We retrieved 13 papers reporting on one or more of the molecules of interest during naturally occurring sleep, 2 during sleep deprivation and 2 during both. Only two studies targeted PFC. Histamine was low during sleep, but high during sleep deprivation and wakefulness, irrespective of brain area. Glu (k = 11) and GABA (k = 8) concentrations in different brain areas were reported to peak during sleep or wakefulness or to lack state-dependency. Aspartate, glycine, asparagine and taurine were less often studied (1-2 times), but peaked exclusively during sleep. Sleep deprivation increased glutamate and GABA exclusively in the cortex. Further studies are needed for drawing solid conclusions.
Collapse
|