1
|
Hristov M, Landzhov B, Yakimova K. Effect of leptin on nitrergic neurons in the lateral hypothalamic area and the supraoptic nucleus of rats. Biotech Histochem 2024; 99:125-133. [PMID: 38533595 DOI: 10.1080/10520295.2024.2335167] [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] [Indexed: 03/28/2024] Open
Abstract
The adipocyte-derived hormone, leptin, plays a key role in the maintenance of energy homeostasis. Leptin binds to the long form of its receptor, which is predominantly expressed in various hypothalamic regions, including the lateral hypothalamic area (LH) and supraoptic nucleus (SO). Several studies have suggested that leptin directly activates neuronal nitric oxide synthase, leading to increased nitric oxide production. We used histochemistry for nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) as a marker for nitric oxide synthase activity and assessed the effect of leptin on nitrergic neurons in the LH and SO of rats. We found that intraperitoneal administration of leptin led to a significant increase in the number of NADPH-d-positive neurons in the LH and SO. In addition, the intensity (optical density) of NADPH-d staining in LH and SO neurons was significantly elevated in rats that received leptin compared with saline-treated rats. These findings suggest that nitrergic neurons in the LH and SO may be implicated in mediating the central effects of leptin.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Krassimira Yakimova
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
2
|
Precocious puberty in narcolepsy type 1: Orexin loss and/or neuroinflammation, which is to blame? Sleep Med Rev 2022; 65:101683. [PMID: 36096986 DOI: 10.1016/j.smrv.2022.101683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 10/14/2022]
Abstract
Narcolepsy type 1 (NT1) is a rare neurological sleep disorder triggered by postnatal loss of the orexin/hypocretin neuropeptides. Overweight/obesity and precocious puberty are highly prevalent comorbidities of NT1, with a close temporal correlation with disease onset, suggesting a common origin. However, the underlying mechanisms remain unknown and merit further investigation. The main question we address in this review is whether the occurrence of precocious puberty in NT1 is due to the lack of orexin/hypocretin or rather to a wider hypothalamic dysfunction in the context of neuroinflammation, which is likely to accompany the disease given its autoimmune origins. Our analysis suggests that the suspected generalized neuroinflammation of the hypothalamus in NT1 would tend to delay puberty rather than hastening it. In contrast, that the brutal loss of orexin/hypocretin would favor an early reactivation of gonadotropin-releasing hormone (GnRH) secretion during the prepubertal period in vulnerable children, leading to early puberty onset. Orexin/hypocretin replacement could thus be envisaged as a potential treatment for precocious puberty in NT1. Additionally, we put forward an alternative hypothesis regarding the concomitant occurrence of sleepiness, weight gain and early puberty in NT1.
Collapse
|
3
|
Kolokolov OV, Salina EA, Yudina VV, Shuldyakov AA, Runnova AE. Infections, Pandemics, and Sleep Disorders. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:319-325. [PMID: 35692961 PMCID: PMC9170495 DOI: 10.1007/s11055-022-01242-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022]
Abstract
Studies of the neurological symptoms and signs associated with the acute and late phases of infectious diseases are important in pandemic conditions. The novel coronavirus infection (COVID-19) pandemic has increased the number of patients with sleeplessness, this being an adverse prognostic factor for infections. This review addresses the factors and mechanisms of sleep impairments and their relationship with inflammation and immune system dysfunction in infectious diseases. In particular, impairments to the functioning of the melatoninergic system are discussed as the cause of sleeplessness during pandemics. The relevance of developing measures for rehabilitating patients, particularly use of Sonnovan to restore normal sleep, which plays a fundamental role in supporting people's mental and physical health, is emphasized.
Collapse
Affiliation(s)
- O V Kolokolov
- Razumovsky Saratov State Medical University, Russian Ministry of Health, Saratov, Russia
| | - E A Salina
- Razumovsky Saratov State Medical University, Russian Ministry of Health, Saratov, Russia
| | - V V Yudina
- Razumovsky Saratov State Medical University, Russian Ministry of Health, Saratov, Russia
| | - A A Shuldyakov
- Razumovsky Saratov State Medical University, Russian Ministry of Health, Saratov, Russia
| | - A E Runnova
- Razumovsky Saratov State Medical University, Russian Ministry of Health, Saratov, Russia
| |
Collapse
|
4
|
Kourosh-Arami M, Hosseini N, Mohsenzadegan M, Komaki A, Joghataei MT. Neurophysiologic implications of neuronal nitric oxide synthase. Rev Neurosci 2021; 31:617-636. [PMID: 32739909 DOI: 10.1515/revneuro-2019-0111] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/21/2020] [Indexed: 12/12/2022]
Abstract
The molecular and chemical properties of neuronal nitric oxide synthase (nNOS) have made it a key mediator in many physiological functions and signaling transduction. The NOS monomer is inactive, but the dimer form is active. There are three forms of NOS, which are neuronal (nNOS), inducible (iNOS), and endothelial (eNOS) nitric oxide synthase. nNOS regulates nitric oxide (NO) synthesis which is the mechanism used mostly by neurons to produce NO. nNOS expression and activation is regulated by some important signaling proteins, such as cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB), calmodulin (CaM), heat shock protein 90 (HSP90)/HSP70. nNOS-derived NO has been implicated in modulating many physiological functions, such as synaptic plasticity, learning, memory, neurogenesis, etc. In this review, we have summarized recent studies that have characterized structural features, subcellular localization, and factors that regulate nNOS function. Finally, we have discussed the role of nNOS in the developing brain under a wide range of physiological conditions, especially long-term potentiation and depression.
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Nasrin Hosseini
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Monireh Mohsenzadegan
- Department of Laboratory Sciences, Allied Medical College, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Alireza Komaki
- Department of Physiology, Medical College, Hamedan University of Medical Sciences, Hamedan, Islamic Republic of Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
5
|
Deurveilher S, Antonchuk M, Saumure BSC, Baldin A, Semba K. No loss of orexin/hypocretin, melanin-concentrating hormone or locus coeruleus noradrenergic neurons in a rat model of chronic sleep restriction. Eur J Neurosci 2021; 54:6027-6043. [PMID: 34355453 DOI: 10.1111/ejn.15412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
Chronic sleep restriction (CSR) is common in modern society, adversely affecting cognitive performance and health. Yet how it impacts neurons regulating sleep remains unclear. Several studies using mice reported substantial losses of wake-active orexin/hypocretin and locus coeruleus (LC) noradrenergic neurons, but not rapid eye movement sleep-active melanin-concentrating hormone (MCH) neurons, following CSR. Here, we used immunohistochemistry and stereology to examine orexin, MCH and LC noradrenergic neurons in a rat model of CSR that uses programmed wheel rotation (3 h on/1 h off; '3/1' protocol). Adult male Wistar rats underwent one or four cycles of the 4-day 3/1 CSR protocol, with 2-day recovery between cycles in home cages. Time-matched control rats were housed in locked wheels/home cages. We found no significant differences in the numbers of orexin, MCH and LC noradrenergic neurons following either one- or four-cycle CSR protocol compared to respective controls. Similarly, the four-cycle CSR protocol had no effect on the densities of orexin axon terminals in the LC, noradrenergic dendrites in the LC and noradrenergic axon terminals in the frontal cortex. Body weights, however, decreased after one cycle of CSR and then increased with diminishing slope over the next three cycles. Thus, we found no evidence for loss of orexin or LC noradrenergic neurons following one and four cycles of the 4-day 3/1 CSR protocol in rats. Differences in CSR protocols and/or possible species differences in neuronal vulnerability to sleep loss may account for the discrepancy between the current results in rats and previous findings in mice.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael Antonchuk
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brock St C Saumure
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Baldin
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
6
|
The Role of Protein S-Nitrosylation in Protein Misfolding-Associated Diseases. Life (Basel) 2021; 11:life11070705. [PMID: 34357077 PMCID: PMC8304259 DOI: 10.3390/life11070705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Abnormal and excessive nitrosative stress contributes to neurodegenerative disease associated with the production of pathological levels of misfolded proteins. The accumulated findings strongly suggest that excessive NO production can induce and deepen these pathological processes, particularly by the S-nitrosylation of target proteins. Therefore, the relationship between S-nitrosylated proteins and the accumulation of misfolded proteins was reviewed. We particularly focused on the S-nitrosylation of E3-ubiquitin-protein ligase, parkin, and endoplasmic reticulum chaperone, PDI, which contribute to the accumulation of misfolded proteins. In addition to the target proteins being S-nitrosylated, NOS, which produces NO, and GSNOR, which inhibits S-nitrosylation, were also suggested as potential therapeutic targets for protein misfolding-associated diseases.
Collapse
|
7
|
Kolokolov OV, Salina EA, Yudina VV, Shuldyakov AA, Runnova AE. [Infections, pandemics and sleep disorders]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:68-74. [PMID: 34078863 DOI: 10.17116/jnevro202112104268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The study of neurological symptoms and signs connected with acute and long-term periods of infectious diseases is relevant during pandemics. The COVID-19 pandemic has increased the number of patients suffering from insomnia. Sleep disturbance is an unfavorable prognostic factor for infections. Risk factors and mechanisms of sleep disturbance, their relationship with inflammation and dysfunction of the immune system in infectious diseases are presented. In particular, dysfunction of the melatonergic system as the cause of insomnia during pandemics is considered. The authors highlight the relevance of measures aimed at the rehabilitation of patients, in particular, the use of sonnovan to restore normal sleep, which is important for a person's mental and physical health.
Collapse
Affiliation(s)
- O V Kolokolov
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - E A Salina
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - V V Yudina
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - A A Shuldyakov
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - A E Runnova
- Razumovsky Saratov State Medical University, Saratov, Russia
| |
Collapse
|
8
|
Kobayashi Y, Oguro A, Yagi E, Mitani A, Kudoh SN, Imaoka S. Bisphenol A and rotenone induce S-nitrosylation of protein disulfide isomerase (PDI) and inhibit neurite outgrowth of primary cultured cells of the rat hippocampus and PC12 cells. J Toxicol Sci 2021; 45:783-794. [PMID: 33268678 DOI: 10.2131/jts.45.783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Bisphenol A (BPA) interferes the function and development of the central nervous system (CNS), resulting in behavioral abnormalities and memory loss. S-nitrosylation of protein disulfide isomerase (PDI) is increased in brains with sporadic Alzheimer's disease and Parkinson's disease. The aim of the present study was to clarify the role of nitric oxide (NO) in BPA-induced neurotoxicity. Since rotenone induces NO-mediated neurodegeneration through S-nitrosylation of PDI, it was used as a positive control. First, rats were treated with BPA and rotenone, and S-nitrosylation of PDI was detected in rat brain microsomes. BPA and rotenone decreased RNase oxidation activity of PDI concomitant with S-nitrosylation of PDI. Next, to clarify S-nitrosylation of PDI by BPA and rotenone in rat brains, we treated the rat pheochromocytoma cell line PC12 and primary cultured neuron cells from the rat hippocampus with BPA (5 and 10 μM) and rotenone (100 or 200 nM). BPA induced S-nitrosylation of PDI, while NG-monomethyl-L-arginine (L-NMMA), a NOS inhibitor, exerted the opposite effects. Finally, to evaluate the toxicity of BPA in the CNS, we investigated its effects on neurite outgrowth of PC12 and primary cultured neuron cells. BPA inhibited neurite outgrowth of these cells, while L-NMMA reversed this inhibition. The involvement of PDI activity in neurite outgrowth was also examined, and bacitracin, a PDI inhibitor, is shown to decrease neurite outgrowth. Furthermore, the overexpression of PDI, but not a catalytically inactive PDI mutant, enhanced neurite outgrowth. These results suggested that S-nitrosylation of PDI induced by excessive NO caused BPA-induced neurotoxicity.
Collapse
Affiliation(s)
- Yukino Kobayashi
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University
| | - Ami Oguro
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University.,Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University
| | - Erina Yagi
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University
| | - Akira Mitani
- Department of Human-System Interaction, School of Science and Technology, Kwansei Gakuin University
| | - Suguru N Kudoh
- Department of Human-System Interaction, School of Science and Technology, Kwansei Gakuin University
| | - Susumu Imaoka
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University
| |
Collapse
|
9
|
PDI-Regulated Disulfide Bond Formation in Protein Folding and Biomolecular Assembly. Molecules 2020; 26:molecules26010171. [PMID: 33396541 PMCID: PMC7794689 DOI: 10.3390/molecules26010171] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Disulfide bonds play a pivotal role in maintaining the natural structures of proteins to ensure their performance of normal biological functions. Moreover, biological molecular assembly, such as the gluten network, is also largely dependent on the intermolecular crosslinking via disulfide bonds. In eukaryotes, the formation and rearrangement of most intra- and intermolecular disulfide bonds in the endoplasmic reticulum (ER) are mediated by protein disulfide isomerases (PDIs), which consist of multiple thioredoxin-like domains. These domains assist correct folding of proteins, as well as effectively prevent the aggregation of misfolded ones. Protein misfolding often leads to the formation of pathological protein aggregations that cause many diseases. On the other hand, glutenin aggregation and subsequent crosslinking are required for the formation of a rheologically dominating gluten network. Herein, the mechanism of PDI-regulated disulfide bond formation is important for understanding not only protein folding and associated diseases, but also the formation of functional biomolecular assembly. This review systematically illustrated the process of human protein disulfide isomerase (hPDI) mediated disulfide bond formation and complemented this with the current mechanism of wheat protein disulfide isomerase (wPDI) catalyzed formation of gluten networks.
Collapse
|
10
|
Błaszczyk JW. Energy Metabolism Decline in the Aging Brain-Pathogenesis of Neurodegenerative Disorders. Metabolites 2020; 10:metabo10110450. [PMID: 33171879 PMCID: PMC7695180 DOI: 10.3390/metabo10110450] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
There is a growing body of evidencethat indicates that the aging of the brain results from the decline of energy metabolism. In particular, the neuronal metabolism of glucose declines steadily, resulting in a growing deficit of adenosine triphosphate (ATP) production-which, in turn, limits glucose access. This vicious circle of energy metabolism at the cellular level is evoked by a rising deficiency of nicotinamide adenine dinucleotide (NAD) in the mitochondrial salvage pathway and subsequent impairment of the Krebs cycle. A decreasing NAD level also impoverishes the activity of NAD-dependent enzymes that augments genetic errors and initiate processes of neuronal degeneration and death.This sequence of events is characteristic of several brain structures in which neurons have the highest energy metabolism. Neurons of the cerebral cortex and basal ganglia with long unmyelinated axons and these with numerous synaptic junctions are particularly prone to senescence and neurodegeneration. Unfortunately, functional deficits of neurodegeneration are initially well-compensated, therefore, clinical symptoms are recognized too late when the damages to the brain structures are already irreversible. Therefore, future treatment strategies in neurodegenerative disorders should focus on energy metabolism and compensation age-related NAD deficit in neurons. This review summarizes the complex interrelationships between metabolic processes on the systemic and cellular levels and provides directions on how to reduce the risk of neurodegeneration and protect the elderly against neurodegenerative diseases.
Collapse
Affiliation(s)
- Janusz Wiesław Błaszczyk
- Department of Human Motor Behavior, Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| |
Collapse
|
11
|
Wibowo E, Garcia ACB, Mainwaring JM. Chronic sleep deprivation prolongs the reduction of sexual behaviour associated with daily sexual encounter in male rats. Physiol Behav 2020; 224:113058. [PMID: 32652091 DOI: 10.1016/j.physbeh.2020.113058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 01/20/2023]
Abstract
Chronic sleep deprivation (CSD) is common in many societies. Consecutive sleep loss increases allostatic load, which is known to negatively affect health outcomes. We investigated the impact of CSD on male sexual behaviour. Sexually-experienced male Long-Evans rats (singly housed under 14:10 light:dark) were either subjected to CSD or no CSD for 7 days, followed by a 7-day sleep recovery (SR) period. Their sexual behaviours were tested daily during both periods. CSD was performed by a 'gentle-handling' protocol for 4 hours per day, at the end of the light phase. Daily sexual behaviour tests led to a change in sexual behaviour over time. Intromission and ejaculation frequencies declined with repeated testing, but the reduction in these behaviours lasted for a longer period in rats that were previously subjected to CSD. Ejaculation latency was significantly longer towards the end of the recovery period in rats that had undergone CSD, but not in the control group. Post-ejaculatory interval increased and mounting behaviour did not change with daily mating tests, regardless of sleep deprivation protocol. CSD prolongs the decline in sexual behaviours associated with daily sexual encounters in male rats, and thus the return to baseline for these parameters requires days.
Collapse
Affiliation(s)
- Erik Wibowo
- Department of Anatomy, University of Otago, Dunedin 9016, New Zealand.
| | - Angela C B Garcia
- Department of Anatomy, University of Otago, Dunedin 9016, New Zealand.
| | | |
Collapse
|
12
|
Yamakawa T, Kurauchi Y, Hisatsune A, Seki T, Katsuki H. Endogenous Nitric Oxide Inhibits, Whereas Awakening Stimuli Increase, the Activity of a Subset of Orexin Neurons. Biol Pharm Bull 2019; 41:1859-1865. [PMID: 30504686 DOI: 10.1248/bpb.b18-00633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lateral hypothalamic area contains neurons expressing neuronal nitric oxide synthase (nNOS), in addition to orexin neurons. Here we examined whether the activity of orexin neurons was regulated by endogenous nitric oxide (NO) in male C57BL/6 mice. Caffeine (30 mg/kg, intraperitoneally (i.p.)) increased the number of orexin neurons positive for c-Fos, a marker of neuronal activity, and also increased the number of NOS/c-Fos-positive cells as identified by reduced nicotinamide adenine dinucleotide phosphate (NADPH) diaphorase histochemistry and c-Fos immunohistochemistry. Diphenhydramine hydrochloride (10 mg/kg. i.p.) decreased c-Fos-positive orexin neurons but had no significant effect on the number of c-Fos-positive NOS neurons. nNOS inhibitor 7-nitroindazole (25 mg/kg, i.p.) alone increased c-Fos-positive orexin neurons, and combined treatment with caffeine and 7-nitroindazole did not show additive effect in the number of c-Fos-positive orexin neurons. In contrast, 7-nitroindazole decreased c-Fos-positive NOS neurons and attenuated caffeine-induced increase in c-Fos-positive NOS neurons. Sleep deprivation increased c-Fos-positive cells in both orexin neurons and NOS neurons, and 7-nitroindazole did not show additive effect with sleep deprivation in the activation of orexin neurons. Together, these results suggest that endogenous NO negatively regulates the activity of a subset of orexin neurons, and this subset of orexin neurons overlaps with that activated by awakening stimuli.
Collapse
Affiliation(s)
- Takao Yamakawa
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Akinori Hisatsune
- Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program," Priority Organization for Innovation and Excellence, Kumamoto University
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
13
|
Alijanpour S, Khakpai F, Ebrahimi-Ghiri M, Zarrindast MR. Co-administration of the low dose of orexin and nitrergic antagonists induces an antidepressant-like effect in mice. Biomed Pharmacother 2019; 109:589-594. [DOI: 10.1016/j.biopha.2018.10.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
|
14
|
|
15
|
Lee DS, Kim JE. PDI-mediated S-nitrosylation of DRP1 facilitates DRP1-S616 phosphorylation and mitochondrial fission in CA1 neurons. Cell Death Dis 2018; 9:869. [PMID: 30158524 PMCID: PMC6115394 DOI: 10.1038/s41419-018-0910-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022]
Abstract
Dynamin-related protein 1 (DRP1) is a key molecule to regulate mitochondrial fission. DRP1 activity is modulated by phosphorylation and S-nitrosylation on serine and cysteine residues, respectively. However, it is still unexplored whether S-nitrosylation of DRP1 affects its phosphorylation. In the present study, we found that Nω-nitro-l-arginine methyl ester hydrochloride (l-NAME, a NOS inhibitor) abolished S-nitrosylated (SNO-DRP1) and DRP1-serine (S) 616 phosphorylation levels in CA1 neurons under physiological condition. l-NAME led to mitochondrial elongation. In spite of the sustained NO synthesis, status epilepticus (a prolonged seizure activity, SE) diminished SNO-DRP1 and DRP1-S616 levels in CA1 neurons, accompanied by the reduced protein disulfide isomerase (PDI) expression and mitochondrial elongation. SE did not influence thioredoxin 1 (Trx1, a denitrosylating enzyme) activity, which was unaffected by l-NAME under physiological and post-SE condition. PDI knockdown decreased SNO-DRP1 and DRP1-S616 levels concomitant with mitochondrial elongation in CA1 neurons without altered NO synthesis under physiological condition. These findings indicate that PDI may be a NO donor of DRP1 to regulate DRP1-S616 phosphorylation, independent of Trx1 activity. Therefore, we suggest that PDI-mediated S-nitrosylation of DRP1 may be one of the major regulatory modifications for mitochondrial dynamics.
Collapse
Affiliation(s)
- Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
16
|
Tesoriero C, Del Gallo F, Bentivoglio M. Sleep and brain infections. Brain Res Bull 2018; 145:59-74. [PMID: 30016726 DOI: 10.1016/j.brainresbull.2018.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022]
Abstract
Sleep is frequently altered in systemic infections as a component of sickness behavior in response to inflammation. Sleepiness in sickness behavior has been extensively investigated. Much less attention has instead been devoted to sleep and wake alterations in brain infections. Most of these, as other neuroinfections, are prevalent in sub-Saharan Africa. The present overview highlights the importance of this topic from both the clinical and pathogenetic points of view. Vigilance states and their regulation are first summarized, emphasizing that key nodes in this distributed brain system can be targeted by neuroinflammatory signaling. Sleep-wake changes in the parasitic disease human African trypanosomiasis (HAT) and its animal models are then reviewed and discussed. Experimental data have revealed that the suprachiasmatic nucleus, the master circadian pacemaker, and peptidergic cell populations of the lateral hypothalamus (the wake-promoting orexin neurons and the sleep-promoting melanin-concentrating hormone neurons) are targeted by African trypanosome infection. It is then discussed how prominent and disturbing are sleep changes in HIV/AIDS, also when the infection is cured with antiretroviral therapy. This recalls attention on the bidirectional interactions between sleep and immune system, including the specialized brain immune response of which microglial cells are protagonists. Sleep changes in an ancient viral disease, rabies, and in the emerging infection due to Zika virus which causes a congenital syndrome, are also dealt with. Altogether the findings indicate that sleep-wake regulation is targeted by brain infections caused by different pathogens and, although the relevant pathogenetic mechanisms largely remain to be clarified, these alterations differ from hypersomnia occurring in sickness behavior. Thus, brain infections point to the vulnerability of the neural network of sleep-wake regulation as a highly relevant clinical and basic science challenge.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Federico Del Gallo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Marina Bentivoglio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy.
| |
Collapse
|
17
|
Laperchia C, Xu YZ, Mumba Ngoyi D, Cotrufo T, Bentivoglio M. Neural Damage in Experimental Trypanosoma brucei gambiense Infection: Hypothalamic Peptidergic Sleep and Wake-Regulatory Neurons. Front Neuroanat 2018. [PMID: 29535612 PMCID: PMC5835115 DOI: 10.3389/fnana.2018.00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuron populations of the lateral hypothalamus which synthesize the orexin (OX)/hypocretin or melanin-concentrating hormone (MCH) peptides play crucial, reciprocal roles in regulating wake stability and sleep. The disease human African trypanosomiasis (HAT), also called sleeping sickness, caused by extracellular Trypanosoma brucei (T. b.) parasites, leads to characteristic sleep-wake cycle disruption and narcoleptic-like alterations of the sleep structure. Previous studies have revealed damage of OX and MCH neurons during systemic infection of laboratory rodents with the non-human pathogenic T. b. brucei subspecies. No information is available, however, on these peptidergic neurons after systemic infection with T. b. gambiense, the etiological agent of 97% of HAT cases. The present study was aimed at the investigation of immunohistochemically characterized OX and MCH neurons after T. b. gambiense or T. b. brucei infection of a susceptible rodent, the multimammate mouse, Mastomysnatalensis. Cell counts and evaluation of OX fiber density were performed at 4 and 8 weeks post-infection, when parasites had entered the brain parenchyma from the periphery. A significant decrease of OX neurons (about 44% reduction) and MCH neurons (about 54% reduction) was found in the lateral hypothalamus and perifornical area at 8 weeks in T. b. gambiense-infected M. natalensis. A moderate decrease (21% and 24% reduction, respectively), which did not reach statistical significance, was found after T. b. brucei infection. In two key targets of diencephalic orexinergic innervation, the peri-suprachiasmatic nucleus (SCN) region and the thalamic paraventricular nucleus (PVT), densitometric analyses showed a significant progressive decrease in the density of orexinergic fibers in both infection paradigms, and especially during T. b. gambiense infection. Altogether the findings provide novel information showing that OX and MCH neurons are highly vulnerable to chronic neuroinflammatory signaling caused by the infection of human-pathogenic African trypanosomes.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Yuan-Zhong Xu
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dieudonné Mumba Ngoyi
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of Congo
| | - Tiziana Cotrufo
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Bentivoglio
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
18
|
Katsuki H. [Cellular mechanisms involved in induction of selective degeneration of orexin neurons in the hypothalamus]. Nihon Yakurigaku Zasshi 2018; 152:70-76. [PMID: 30101863 DOI: 10.1254/fpj.152.70] [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: 06/08/2023]
Abstract
Narcolepsy is a kind of sleep disorder featured by selective loss of orexin neurons in the lateral hypothalamus. Several lines of evidence, including association with specific HLA haplotypes, gene polymorphism in T cell receptor and detection of autoantibodies in a subpopulation of patients, suggest that autoimmune responses play an important role in the pathogenesis of this disorder. Potential relationship with influenza virus infection has also been a matter of interest. However, these events may not be able to explain all cases of narcolepsy. Based on the structural features of orexin, in addition to the findings on the characteristics of orexin neurons obtained from studies in organotypic hypothalamic slice cultures, we proposed novel mechanisms potentially involved in selective degeneration of orexin neurons. Increase in local production of nitric oxide induced by several life style-related conditions such as shortage of sleep and intake of high fat diet leads to inactivation of protein disulfide isomerase. Consequently, abnormal aggregates of orexin and/or its precursor that possess two intra-molecular disulfide bonds accumulate within orexin neurons. In addition to the increase in endoplasmic reticulum stress, accumulation of orexin as abnormal aggregates leads to increased excitability of orexin neurons by shutdown of feedback inhibition resulting from deficits in orexin release. These mechanisms may provide a clue to understand the pathogenic mechanisms of various neurological and psychiatric disorders accompanied by a decrease of orexin.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
19
|
Wang YW, Zhou Q, Zhang X, Qian QQ, Xu JW, Ni PF, Qian YN. Mild endoplasmic reticulum stress ameliorates lipopolysaccharide-induced neuroinflammation and cognitive impairment via regulation of microglial polarization. J Neuroinflammation 2017; 14:233. [PMID: 29179727 PMCID: PMC5704515 DOI: 10.1186/s12974-017-1002-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neuroinflammation, which ultimately leads to neuronal loss, is considered to play a crucial role in numerous neurodegenerative diseases. The neuroinflammatory process is characterized by the activation of glial cells such as microglia. Endoplasmic reticulum (ER) stress is commonly associated with impairments in neuronal function and cognition, but its relationship and role in neurodegeneration is still controversial. Recently, it was confirmed that nonharmful levels of ER stress protected against experimental Parkinson's disease. Here, we investigated mild ER stress-based regulation of lipopolysaccharide (LPS)-driven neuroinflammation in rats and in primary microglia. METHODS Male Sprague-Dawley (SD) rats received the intracerebroventricular injection of the ER stress activator tunicamycin (TM) with or without intraperitoneal injection of the ER stress stabilizer sodium 4-phenylbutyrate (4-PBA) 1 h before LPS administration. The levels of neuroinflammation and memory dysfunction were assessed 24 h after treatment. In addition, the effect of mild ER stress on microglia was determined in vitro. RESULTS Here, we found that low doses of TM led to mild ER stress without cell or organism lethality. We showed that mild ER stress preconditioning reduced microglia activation and neuronal death as well as improved LPS-induced memory impairment in rats. In addition, pre-exposure to nonlethal doses of TM in microglia showed significant protection against LPS-induced proinflammatory cytokine production and M1/2b polarization. However, sodium 4-PBA, a compound that ameliorates ER stress, ablated this protective effect in vivo and in vitro. CONCLUSIONS Based on our findings, we conclude that the mild ER stress not only limits the accumulation of misfolded proteins but also protects tissues from harmful endotoxemia insults. Therefore, ER stress preconditioning has potential therapeutic value for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Wei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Qin Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiang Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Qing-Qing Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Jia-Wen Xu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Peng-Fei Ni
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yan-Ning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China.
| |
Collapse
|
20
|
Zhu X, Dong J, Han B, Huang R, Zhang A, Xia Z, Chang H, Chao J, Yao H. Neuronal Nitric Oxide Synthase Contributes to PTZ Kindling Epilepsy-Induced Hippocampal Endoplasmic Reticulum Stress and Oxidative Damage. Front Cell Neurosci 2017; 11:377. [PMID: 29234274 PMCID: PMC5712337 DOI: 10.3389/fncel.2017.00377] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022] Open
Abstract
Epilepsy is one of the most common chronic neurological disorders which provoke progressive neuronal degeneration. Endoplasmic reticulum (ER) stress has recently been recognized as pivotal etiological factors contributing to epilepsy-induced neuronal damage. However, the specific contribution of epilepsy made to ER stress remains largely elusive. Here we use pentylenetetrazole (PTZ) kindling, a chronic epilepsy model, to identify neuronal nitric oxide synthase (nNOS) as a signaling molecule triggering PTZ kindling epilepsy-induced ER stress and oxidative damage. By genetic deletion of nNOS gene, we further demonstrated that nNOS acts through peroxynitrite, an important member of reactive nitrogen species, to trigger hippocampal ER stress and oxidative damage in the PTZ-kindled mice. Our findings thus define a specific mechanism for chronic epilepsy-induced ER stress and oxidative damage, and identify a potential therapeutic target for neuroprotection in chronic epilepsy patients.
Collapse
Affiliation(s)
- Xinjian Zhu
- Department of Pharmacology, Medical School, Southeast University, Nanjing, China
| | - Jingde Dong
- Department of Geriatric Neurology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Bing Han
- Department of Pharmacology, Medical School, Southeast University, Nanjing, China
| | - Rongrong Huang
- Department of Pharmacology, Medical School, Southeast University, Nanjing, China
| | - Aifeng Zhang
- Department of Pathology, Medical School, Southeast University, Nanjing, China
| | - Zhengrong Xia
- Analysis and Test Center, Nanjing Medical University, Nanjing, China
| | - Huanhuan Chang
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Jie Chao
- Department of Physiology, Medical School, Southeast University, Nanjing, China
| | - Honghong Yao
- Department of Pharmacology, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
21
|
Carrier J, Semba K, Deurveilher S, Drogos L, Cyr-Cronier J, Lord C, Sekerovick Z. Sex differences in age-related changes in the sleep-wake cycle. Front Neuroendocrinol 2017; 47:66-85. [PMID: 28757114 DOI: 10.1016/j.yfrne.2017.07.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/09/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
Age-related changes in sleep and circadian regulation occur as early as the middle years of life. Research also suggests that sleep and circadian rhythms are regulated differently between women and men. However, does sleep and circadian rhythms regulation age similarly in men and women? In this review, we present the mechanisms underlying age-related differences in sleep and the current state of knowledge on how they interact with sex. We also address how testosterone, estrogens, and progesterone fluctuations across adulthood interact with sleep and circadian regulation. Finally, we will propose research avenues to unravel the mechanisms underlying sex differences in age-related effects on sleep.
Collapse
Affiliation(s)
- Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada; Institut Universitaire de Gériatrie de Montréal, Université de Montréal, Montréal, Québec, Canada; Département de psychologie, Université de Montréal, Montréal, Québec, Canada.
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lauren Drogos
- Departments of Physiology & Pharmacology and Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Jessica Cyr-Cronier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| | - Catherine Lord
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| | - Zoran Sekerovick
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
22
|
Diurnal fluctuation in the number of hypocretin/orexin and histamine producing: Implication for understanding and treating neuronal loss. PLoS One 2017; 12:e0178573. [PMID: 28570646 PMCID: PMC5453544 DOI: 10.1371/journal.pone.0178573] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/15/2017] [Indexed: 11/24/2022] Open
Abstract
The loss of specific neuronal phenotypes, as determined by immunohistochemistry, has become a powerful tool for identifying the nature and cause of neurological diseases. Here we show that the number of neurons identified and quantified using this method misses a substantial percentage of extant neurons in a phenotype specific manner. In mice, 24% more hypocretin/orexin (Hcrt) neurons are seen in the night compared to the day, and an additional 17% are seen after inhibiting microtubule polymerization with colchicine. We see no such difference between the number of MCH (melanin concentrating hormone) neurons in dark, light or colchicine conditions, despite MCH and Hcrt both being hypothalamic peptide transmitters. Although the size of Hcrt neurons did not differ between light and dark, the size of MCH neurons was increased by 15% in the light phase. The number of neurons containing histidine decarboxylase (HDC), the histamine synthesizing enzyme, was 34% greater in the dark than in the light, but, like Hcrt, cell size did not differ. We did not find a significant difference in the number or the size of neurons expressing choline acetyltransferase (ChAT), the acetylcholine synthesizing enzyme, in the horizontal diagonal band (HBD) during the dark and light conditions. As expected, colchicine treatment did not increase the number of these neurons. Understanding the function and dynamics of transmitter production within “non-visible” phenotypically defined cells has fundamental implications for our understanding of brain plasticity.
Collapse
|
23
|
Fujita I, Nobunaga M, Seki T, Kurauchi Y, Hisatsune A, Katsuki H. Cystamine-mediated inhibition of protein disulfide isomerase triggers aggregation of misfolded orexin-A in the Golgi apparatus and prevents extracellular secretion of orexin-A. Biochem Biophys Res Commun 2017; 489:164-170. [PMID: 28549585 DOI: 10.1016/j.bbrc.2017.05.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/22/2017] [Indexed: 10/19/2022]
Abstract
Orexins (orexin-A and orexin-B) are neuropeptides that are reduced in narcolepsy, a sleep disorder that is characterized by excessive daytime sleepiness, sudden sleep attacks and cataplexy. However, it remains unclear how orexins in the brain and orexin neurons are reduced in narcolepsy. Orexin-A has two closely located intramolecular disulfide bonds and is prone to misfolding due to the formation of incorrect disulfide bonds. Protein disulfide isomerase (PDI) possesses disulfide interchange activity. PDI can modify misfolded orexin-A to its native form by rearrangement of two disulfide bonds. We have previously demonstrated that sleep deprivation and a high fat diet increase nitric oxide in the brain. This increase triggers S-nitrosation and inactivation of PDI, leading to aggregation of orexin-A and reduction of orexin neurons. However, the relationship between PDI inactivation and loss of orexin neurons has not yet been fully elucidated. In the present study, we used a PDI inhibitor, cystamine, to elucidate the precise molecular mechanism by which PDI inhibition reduces the number of orexin neurons. In rat hypothalamic slice cultures, cystamine induced selective depletion of orexin-A, but not orexin-B and melanin-concentrating hormone. Moreover, cystamine triggered aggregation of orexin-A, but not orexin-B in the Golgi apparatus of hypothalamic slice cultures and in vivo mouse brains. However, cystamine did not induce endoplasmic reticulum (ER) stress, and an ER stress inducer did not trigger aggregation of orexin-A in slice cultures. Finally, we demonstrated that cystamine significantly decreased extracellular secretion of orexin-A in AD293 cells overexpressing prepro-orexin. These findings suggest that cystamine-induced PDI inhibition induces selective depletion, aggregation in the Golgi apparatus and impaired secretion of orexin-A. These effects may represent an initial step in the pathogenesis of narcolepsy.
Collapse
Affiliation(s)
- Issei Fujita
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mizuki Nobunaga
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akinori Hisatsune
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Japan; Program for Leading Graduate Schools "HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program", Kumamoto University, Kumamoto, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
24
|
Kamali AM, Noorafshan A, Karimi F, Karbalay-Doust S, Nami M. The Impact of Chronic Sleep Restriction on Neuronal Number and Volumetric Correlates of the Dorsal Respiratory Nuclei in a Rat Model. Sleep 2017; 40:3787185. [DOI: 10.1093/sleep/zsx072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
25
|
Laperchia C, Palomba M, Seke Etet PF, Rodgers J, Bradley B, Montague P, Grassi-Zucconi G, Kennedy PGE, Bentivoglio M. Trypanosoma brucei Invasion and T-Cell Infiltration of the Brain Parenchyma in Experimental Sleeping Sickness: Timing and Correlation with Functional Changes. PLoS Negl Trop Dis 2016; 10:e0005242. [PMID: 28002454 PMCID: PMC5217973 DOI: 10.1371/journal.pntd.0005242] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/06/2017] [Accepted: 12/07/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The timing of Trypanosoma brucei entry into the brain parenchyma to initiate the second, meningoencephalitic stage of human African trypanosomiasis or sleeping sickness is currently debated and even parasite invasion of the neuropil has been recently questioned. Furthermore, the relationship between neurological features and disease stage are unclear, despite the important diagnostic and therapeutic implications. METHODOLOGY Using a rat model of chronic Trypanosoma brucei brucei infection we determined the timing of parasite and T-cell neuropil infiltration and its correlation with functional changes. Parasite DNA was detected using trypanosome-specific PCR. Body weight and sleep structure alterations represented by sleep-onset rapid eye movement (SOREM) periods, reported in human and experimental African trypanosomiasis, were monitored. The presence of parasites, as well as CD4+ and CD8+ T-cells in the neuropil was assessed over time in the brain of the same animals by immunocytochemistry and quantitative analyses. PRINCIPAL FINDINGS Trypanosome DNA was present in the brain at day 6 post-infection and increased more than 15-fold by day 21. Parasites and T-cells were observed in the parenchyma from day 9 onwards. Parasites traversing blood vessel walls were observed in the hypothalamus and other brain regions. Body weight gain was reduced from day 7 onwards. SOREM episodes started in most cases early after infection, with an increase in number and duration after parasite neuroinvasion. CONCLUSION These findings demonstrate invasion of the neuropil over time, after an initial interval, by parasites and lymphocytes crossing the blood-brain barrier, and show that neurological features can precede this event. The data thus challenge the current clinical and cerebrospinal fluid criteria of disease staging.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maria Palomba
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Paul F. Seke Etet
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Jean Rodgers
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Barbara Bradley
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Paul Montague
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Gigliola Grassi-Zucconi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Peter G. E. Kennedy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
26
|
Hunt NJ, Waters KA, Machaalani R. Promotion of the Unfolding Protein Response in Orexin/Dynorphin Neurons in Sudden Infant Death Syndrome (SIDS): Elevated pPERK and ATF4 Expression. Mol Neurobiol 2016; 54:7171-7185. [PMID: 27796753 DOI: 10.1007/s12035-016-0234-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/16/2016] [Indexed: 01/08/2023]
Abstract
We previously demonstrated that sudden infant death syndrome (SIDS) infants have decreased orexin immunoreactivity within the hypothalamus and pons compared to non-SIDS infants. In this study, we examined multiple mechanisms that may promote loss of orexin expression including programmed cell death, impaired maturation/structural stability, neuroinflammation and impaired unfolding protein response (UPR). Immunofluorescent and immunohistochemical staining for a number of markers was performed in the tuberal hypothalamus and pons of infants (1-10 months) who died from SIDS (n = 27) compared to age- and sex-matched non-SIDS infants (n = 19). The markers included orexin A (OxA), dynorphin (Dyn), cleaved caspase 3 (CC3), cleaved caspase 9 (CC9), glial fibrillary acid protein (GFAP), tubulin beta chain 3 (TUBB3), myelin basic protein (MBP), interleukin 1β (IL-1β), terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL), c-fos and the UPR activation markers: phosphorylated protein kinase RNA-like endoplasmic reticulum kinase (pPERK), and activating transcription factor 4 (ATF4). It was hypothesised that pPERK and ATF4 would be upregulated in Ox neurons in SIDS compared to non-SIDS. Within the hypothalamus, OxA and Dyn co-localised with a 20 % decrease in expression in SIDS infants (P = 0.001). pPERK and ATF4 expression in OxA neurons were increased by 35 % (P = 0.001) and 15 % (P = 0.001) respectively, with linear relationships between the decreased OxA/Dyn expression and the percentages of co-localised pPERK/OxA and ATF4/OxA evident (P = 0.01, P = 0.01). No differences in co-localisation with CC9, CC3, TUNEL or c-fos, nor expression of MBP, TUBB3, IL-1β and GFAP, were observed in the hypothalamus. In the pons, there were 40 % and 20 % increases in pPERK expression in the locus coeruleus (P = 0.001) and dorsal raphe (P = 0.022) respectively; ATF4 expression was not changed. The findings that decreased orexin levels in SIDS infants may be associated with an accumulation of pPERK suggest decreased orexin translation. As pPERK may inhibit multiple neuronal groups in the pons in SIDS infants, it could also indicate that a common pathway promotes loss of protein expression and impaired functionality of multiple brainstem neuronal groups.
Collapse
Affiliation(s)
- Nicholas J Hunt
- SIDS and Sleep Apnoea Laboratory, Department of Medicine, Sydney Medical School, University of Sydney, Room 206, Blackburn Building, D06, Sydney, NSW, Australia.,BOSCH Institute of Biomedical Research, University of Sydney, Sydney, NSW, Australia
| | - Karen A Waters
- SIDS and Sleep Apnoea Laboratory, Department of Medicine, Sydney Medical School, University of Sydney, Room 206, Blackburn Building, D06, Sydney, NSW, Australia.,BOSCH Institute of Biomedical Research, University of Sydney, Sydney, NSW, Australia.,The Children's Hospital, Westmead, NSW, Australia
| | - Rita Machaalani
- SIDS and Sleep Apnoea Laboratory, Department of Medicine, Sydney Medical School, University of Sydney, Room 206, Blackburn Building, D06, Sydney, NSW, Australia. .,BOSCH Institute of Biomedical Research, University of Sydney, Sydney, NSW, Australia. .,The Children's Hospital, Westmead, NSW, Australia.
| |
Collapse
|
27
|
Zhu Y, Fenik P, Zhan G, Somach R, Xin R, Veasey S. Intermittent Short Sleep Results in Lasting Sleep Wake Disturbances and Degeneration of Locus Coeruleus and Orexinergic Neurons. Sleep 2016; 39:1601-11. [PMID: 27306266 DOI: 10.5665/sleep.6030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/11/2016] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Intermittent short sleep (ISS) is pervasive among students and workers in modern societies, yet the lasting consequences of repeated short sleep on behavior and brain health are largely unexplored. Wake-activated neurons may be at increased risk of metabolic injury across sustained wakefulness. METHODS To examine the effects of ISS on wake-activated neurons and wake behavior, wild-type mice were randomized to ISS (a repeated pattern of short sleep on 3 consecutive days followed by 4 days of recovery sleep for 4 weeks) or rested control conditions. Subsets of both groups were allowed a recovery period consisting of 4-week unperturbed activity in home cages with littermates. Mice were examined for immediate and delayed (following recovery) effects of ISS on wake neuron cell metabolics, cell counts, and sleep/wake patterns. RESULTS ISS resulted in sustained disruption of sleep/wake activity, with increased wakefulness during the lights-on period and reduced wake bout duration and wake time during the lights-off period. Noradrenergic locus coeruleus (LC) and orexinergic neurons showed persistent alterations in morphology, and reductions in both neuronal stereological cell counts and fronto-cortical projections. Surviving wake-activated neurons evidenced persistent reductions in sirtuins 1 and 3 and increased lipofuscin. In contrast, ISS resulted in no lasting injury to the sleep-activated melanin concentrating hormone neurons. CONCLUSIONS Collectively these findings demonstrate for the first time that ISS imparts significant lasting disturbances in sleep/wake activity, degeneration of wake-activated LC and orexinergic neurons, and lasting metabolic changes in remaining neurons most consistent with premature senescence.
Collapse
Affiliation(s)
- Yan Zhu
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| | - Guanxia Zhan
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| | - Rebecca Somach
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| | - Ryan Xin
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| | - Sigrid Veasey
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA
| |
Collapse
|
28
|
Cutler RG, Camandola S, Malott KF, Edelhauser MA, Mattson MP. The Role of Uric Acid and Methyl Derivatives in the Prevention of Age-Related Neurodegenerative Disorders. Curr Top Med Chem 2016; 15:2233-8. [PMID: 26059354 DOI: 10.2174/1568026615666150610143234] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/13/2015] [Accepted: 05/04/2015] [Indexed: 12/13/2022]
Abstract
High uric acid (UA levels have been correlated with a reduced risk of many neurodegenerative diseases through mechanisms involving chelating Fenton reaction transitional metals, antioxidant quenching of superoxide and hydroxyl free radicals, and as an electron donor that increases antioxidant enzyme activity (e.g. SOD. However, the clinical usefulness of UA is limited by its' low water solubility and propensity to form inflammatory crystals at hyperuricemic levels. This review focuses on the role of UA in neuroprotection, as well as potential strategies aimed at increasing UA levels in the soluble range, and the potential therapeutic use of more water-soluble methyl-UA derivatives from the natural catabolic end-products of dietary caffeine, theophylline, and theobromine.
Collapse
Affiliation(s)
- Roy G Cutler
- Laboratory of Neurosciences, 251 Bayview Blvd, Baltimore, MD, 21224, U.S.A.
| | | | | | | | | |
Collapse
|
29
|
Wellhauser L, Chalmers JA, Belsham DD. Nitric Oxide Exerts Basal and Insulin-Dependent Anorexigenic Actions in POMC Hypothalamic Neurons. Mol Endocrinol 2016; 30:402-16. [PMID: 26930171 DOI: 10.1210/me.2015-1275] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The arcuate nucleus of the hypothalamus represents a key center for the control of appetite and feeding through the regulation of 2 key neuronal populations, notably agouti-related peptide/neuropeptide Y and proopimelanocortin (POMC)/cocaine- and amphetamine-regulated transcript neurons. Altered regulation of these neuronal networks, in particular the dysfunction of POMC neurons upon high-fat consumption, is a major pathogenic mechanism involved in the development of obesity and type 2 diabetes mellitus. Efforts are underway to preserve the integrity or enhance the functionality of POMC neurons in order to prevent or treat these metabolic diseases. Here, we report for the first time that the nitric oxide (NO(-)) donor, sodium nitroprusside (SNP) mediates anorexigenic actions in both hypothalamic tissue and hypothalamic-derived cell models by mediating the up-regulation of POMC levels. SNP increased POMC mRNA in a dose-dependent manner and enhanced α-melanocortin-secreting hormone production and secretion in mHypoA-POMC/GFP-2 cells. SNP also enhanced insulin-driven POMC expression likely by inhibiting the deacetylase activity of sirtuin 1. Furthermore, SNP enhanced insulin-dependent POMC expression, likely by reducing the transcriptional repression of Foxo1 on the POMC gene. Prolonged SNP exposure prevented the development of insulin resistance. Taken together, the NO(-) donor SNP enhances the anorexigenic potential of POMC neurons by promoting its transcriptional expression independent and in cooperation with insulin. Thus, increasing cellular NO(-) levels represents a hormone-independent method of promoting anorexigenic output from the existing POMC neuronal populations and may be advantageous in the fight against these prevalent disorders.
Collapse
Affiliation(s)
- Leigh Wellhauser
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Jennifer A Chalmers
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Denise D Belsham
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
30
|
H1N1 influenza virus induces narcolepsy-like sleep disruption and targets sleep-wake regulatory neurons in mice. Proc Natl Acad Sci U S A 2015; 113:E368-77. [PMID: 26668381 DOI: 10.1073/pnas.1521463112] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
An increased incidence in the sleep-disorder narcolepsy has been associated with the 2009-2010 pandemic of H1N1 influenza virus in China and with mass vaccination campaigns against influenza during the pandemic in Finland and Sweden. Pathogenetic mechanisms of narcolepsy have so far mainly focused on autoimmunity. We here tested an alternative working hypothesis involving a direct role of influenza virus infection in the pathogenesis of narcolepsy in susceptible subjects. We show that infection with H1N1 influenza virus in mice that lack B and T cells (Recombinant activating gene 1-deficient mice) can lead to narcoleptic-like sleep-wake fragmentation and sleep structure alterations. Interestingly, the infection targeted brainstem and hypothalamic neurons, including orexin/hypocretin-producing neurons that regulate sleep-wake stability and are affected in narcolepsy. Because changes occurred in the absence of adaptive autoimmune responses, the findings show that brain infections with H1N1 virus have the potential to cause per se narcoleptic-like sleep disruption.
Collapse
|
31
|
Qu Z, Greenlief CM, Gu Z. Quantitative Proteomic Approaches for Analysis of Protein S-Nitrosylation. J Proteome Res 2015; 15:1-14. [DOI: 10.1021/acs.jproteome.5b00857] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | - C. Michael Greenlief
- Department
of Chemistry, University of Missouri College of Arts and Science, Columbia, Missouri 65211, United States
| | - Zezong Gu
- Harry S. Truman Veterans’ Hospital, Columbia, Missouri 65201, United States
| |
Collapse
|
32
|
Hunt NJ, Waters KA, Rodriguez ML, Machaalani R. Decreased orexin (hypocretin) immunoreactivity in the hypothalamus and pontine nuclei in sudden infant death syndrome. Acta Neuropathol 2015; 130:185-98. [PMID: 25953524 DOI: 10.1007/s00401-015-1437-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 11/29/2022]
Abstract
Infants at risk of sudden infant death syndrome (SIDS) have been shown to have dysfunctional sleep and poor arousal thresholds. In animal studies, both these attributes have been linked to impaired signalling of the neuropeptide orexin. This study examined the immunoreactivity of orexin (OxA and OxB) in the tuberal hypothalamus (n = 27) and the pons (n = 15) of infants (1-10 months) who died from SIDS compared to age-matched non-SIDS infants. The percentage of orexin immunoreactive neurons and the total number of neurons were quantified in the dorsomedial, perifornical and lateral hypothalamus at three levels of the tuberal hypothalamus. In the pons, the area of orexin immunoreactive fibres were quantified in the locus coeruleus (LC), dorsal raphe (DR), laterodorsal tegmental (LDT), medial parabrachial, dorsal tegmental (DTg) and pontine nuclei (Pn) using automated methods. OxA and OxB were co-expressed in all hypothalamic and pontine nuclei examined. In SIDS infants, orexin immunoreactivity was decreased by up to 21 % within each of the three levels of the hypothalamus compared to non-SIDS (p ≤ 0.050). In the pons, a 40-50 % decrease in OxA occurred in the all pontine nuclei, while a similar decrease in OxB immunoreactivity was observed in the LC, LDT, DTg and Pn (p ≤ 0.025). No correlations were found between the decreased orexin immunoreactivity and previously identified risk factors for SIDS, including prone sleeping position and cigarette smoke exposure. This finding of reduced orexin immunoreactivity in SIDS infants may be associated with sleep dysfunction and impaired arousal.
Collapse
Affiliation(s)
- Nicholas J Hunt
- Department of Medicine, Room 206, SIDS and Sleep Apnoea Laboratory, Sydney Medical School, University of Sydney, Blackburn Building, D06, Sydney, NSW, 2006, Australia
| | | | | | | |
Collapse
|
33
|
Lucke-Wold BP, Smith KE, Nguyen L, Turner RC, Logsdon AF, Jackson GJ, Huber JD, Rosen CL, Miller DB. Sleep disruption and the sequelae associated with traumatic brain injury. Neurosci Biobehav Rev 2015; 55:68-77. [PMID: 25956251 PMCID: PMC4721255 DOI: 10.1016/j.neubiorev.2015.04.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/17/2015] [Accepted: 04/25/2015] [Indexed: 02/08/2023]
Abstract
Sleep disruption, which includes a loss of sleep as well as poor quality fragmented sleep, frequently follows traumatic brain injury (TBI) impacting a large number of patients each year in the United States. Fragmented and/or disrupted sleep can worsen neuropsychiatric, behavioral, and physical symptoms of TBI. Additionally, sleep disruption impairs recovery and can lead to cognitive decline. The most common sleep disruption following TBI is insomnia, which is difficulty staying asleep. The consequences of disrupted sleep following injury range from deranged metabolomics and blood brain barrier compromise to altered neuroplasticity and degeneration. There are several theories for why sleep is necessary (e.g., glymphatic clearance and metabolic regulation) and these may help explain how sleep disruption contributes to degeneration within the brain. Experimental data indicate disrupted sleep allows hyperphosphorylated tau and amyloid β plaques to accumulate. As sleep disruption may act as a cellular stressor, target areas warranting further scientific investigation include the increase in endoplasmic reticulum and oxidative stress following acute periods of sleep deprivation. Potential treatment options for restoring the normal sleep cycle include melatonin derivatives and cognitive behavioral therapy.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Kelly E Smith
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA; The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Linda Nguyen
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA; The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Ryan C Turner
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Aric F Logsdon
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA; The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Garrett J Jackson
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Jason D Huber
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA; The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Charles L Rosen
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Diane B Miller
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; Centers for Disease Control and Prevention-National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
34
|
Shan L, Dauvilliers Y, Siegel JM. Interactions of the histamine and hypocretin systems in CNS disorders. Nat Rev Neurol 2015; 11:401-13. [PMID: 26100750 DOI: 10.1038/nrneurol.2015.99] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Histamine and hypocretin neurons are localized to the hypothalamus, a brain area critical to autonomic function and sleep. Narcolepsy type 1, also known as narcolepsy with cataplexy, is a neurological disorder characterized by excessive daytime sleepiness, impaired night-time sleep, cataplexy, sleep paralysis and short latency to rapid eye movement (REM) sleep after sleep onset. In narcolepsy, 90% of hypocretin neurons are lost; in addition, two groups reported in 2014 that the number of histamine neurons is increased by 64% or more in human patients with narcolepsy, suggesting involvement of histamine in the aetiology of this disorder. Here, we review the role of the histamine and hypocretin systems in sleep-wake modulation. Furthermore, we summarize the neuropathological changes to these two systems in narcolepsy and discuss the possibility that narcolepsy-associated histamine abnormalities could mediate or result from the same processes that cause the hypocretin cell loss. We also review the changes in the hypocretin and histamine systems, and the associated sleep disruptions, in Parkinson disease, Alzheimer disease, Huntington disease and Tourette syndrome. Finally, we discuss novel therapeutic approaches for manipulation of the histamine system.
Collapse
Affiliation(s)
- Ling Shan
- Department of Psychiatry and Brain Research Institute, UCLA School of Medicine, Veterans' Affairs Greater Los Angeles Healthcare System (VA GLAHS), 16111 Plummer Street North Hills, 151A3, CA 91343, USA
| | - Yves Dauvilliers
- Centre de Référence Nationale Maladies Rares, Narcolepsie et Hypersomnie Idiopathique, Département de Neurologie, Hôpital Gui-de-Chauliac, INSERM U1061, 80 avenue Augustin Fliche, Montpellier 34295, France
| | - Jerome M Siegel
- Department of Psychiatry and Brain Research Institute, UCLA School of Medicine, Veterans' Affairs Greater Los Angeles Healthcare System (VA GLAHS), 16111 Plummer Street North Hills, 151A3, CA 91343, USA
| |
Collapse
|
35
|
Nakamura T, Prikhodko OA, Pirie E, Nagar S, Akhtar MW, Oh CK, McKercher SR, Ambasudhan R, Okamoto SI, Lipton SA. Aberrant protein S-nitrosylation contributes to the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2015; 84:99-108. [PMID: 25796565 DOI: 10.1016/j.nbd.2015.03.017] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/09/2015] [Accepted: 03/12/2015] [Indexed: 11/29/2022] Open
Abstract
Nitric oxide (NO) is a gasotransmitter that impacts fundamental aspects of neuronal function in large measure through S-nitrosylation, a redox reaction that occurs on regulatory cysteine thiol groups. For instance, S-nitrosylation regulates enzymatic activity of target proteins via inhibition of active site cysteine residues or via allosteric regulation of protein structure. During normal brain function, protein S-nitrosylation serves as an important cellular mechanism that modulates a diverse array of physiological processes, including transcriptional activity, synaptic plasticity, and neuronal survival. In contrast, emerging evidence suggests that aging and disease-linked environmental risk factors exacerbate nitrosative stress via excessive production of NO. Consequently, aberrant S-nitrosylation occurs and represents a common pathological feature that contributes to the onset and progression of multiple neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. In the current review, we highlight recent key findings on aberrant protein S-nitrosylation showing that this reaction triggers protein misfolding, mitochondrial dysfunction, transcriptional dysregulation, synaptic damage, and neuronal injury. Specifically, we discuss the pathological consequences of S-nitrosylated parkin, myocyte enhancer factor 2 (MEF2), dynamin-related protein 1 (Drp1), protein disulfide isomerase (PDI), X-linked inhibitor of apoptosis protein (XIAP), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) under neurodegenerative conditions. We also speculate that intervention to prevent these aberrant S-nitrosylation events may produce novel therapeutic agents to combat neurodegenerative diseases.
Collapse
Affiliation(s)
- Tomohiro Nakamura
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Olga A Prikhodko
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Elaine Pirie
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Saumya Nagar
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mohd Waseem Akhtar
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Chang-Ki Oh
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Scott R McKercher
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Rajesh Ambasudhan
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shu-ichi Okamoto
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Neurosciences, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
36
|
Tunicamycin-induced unfolded protein response in the developing mouse brain. Toxicol Appl Pharmacol 2015; 283:157-67. [PMID: 25620058 DOI: 10.1016/j.taap.2014.12.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/29/2014] [Accepted: 12/05/2014] [Indexed: 12/23/2022]
Abstract
Accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) causes ER stress, resulting in the activation of the unfolded protein response (UPR). ER stress and UPR are associated with many neurodevelopmental and neurodegenerative disorders. The developing brain is particularly susceptible to environmental insults which may cause ER stress. We evaluated the UPR in the brain of postnatal mice. Tunicamycin, a commonly used ER stress inducer, was administered subcutaneously to mice of postnatal days (PDs) 4, 12 and 25. Tunicamycin caused UPR in the cerebral cortex, hippocampus and cerebellum of mice of PD4 and PD12, which was evident by the upregulation of ATF6, XBP1s, p-eIF2α, GRP78, GRP94 and MANF, but failed to induce UPR in the brain of PD25 mice. Tunicamycin-induced UPR in the liver was observed at all stages. In PD4 mice, tunicamycin-induced caspase-3 activation was observed in layer II of the parietal and optical cortex, CA1-CA3 and the subiculum of the hippocampus, the cerebellar external germinal layer and the superior/inferior colliculus. Tunicamycin-induced caspase-3 activation was also shown on PD12 but to a much lesser degree and mainly located in the dentate gyrus of the hippocampus, deep cerebellar nuclei and pons. Tunicamycin did not activate caspase-3 in the brain of PD25 mice and the liver of all stages. Similarly, immature cerebellar neurons were sensitive to tunicamycin-induced cell death in culture, but became resistant as they matured in vitro. These results suggest that the UPR is developmentally regulated and the immature brain is more susceptible to ER stress.
Collapse
|
37
|
Stern AL, Naidoo N. Wake-active neurons across aging and neurodegeneration: a potential role for sleep disturbances in promoting disease. SPRINGERPLUS 2015; 4:25. [PMID: 25635245 PMCID: PMC4306674 DOI: 10.1186/s40064-014-0777-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/23/2014] [Indexed: 12/13/2022]
Abstract
Sleep/wake disturbance is a feature of almost all common age-related neurodegenerative diseases. Although the reason for this is unknown, it is likely that this inability to maintain sleep and wake states is in large part due to declines in the number and function of wake-active neurons, populations of cells that fire only during waking and are silent during sleep. Consistent with this, many of the brain regions that are most susceptible to neurodegeneration are those that are necessary for wake maintenance and alertness. In the present review, these wake-active populations are systematically assessed in terms of their observed pathology across aging and several neurodegenerative diseases, with implications for future research relating sleep and wake disturbances to aging and age-related neurodegeneration.
Collapse
Affiliation(s)
- Anna L Stern
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Nirinjini Naidoo
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
38
|
Palomba M, Seke-Etet PF, Laperchia C, Tiberio L, Xu YZ, Colavito V, Grassi-Zucconi G, Bentivoglio M. Alterations of orexinergic and melanin-concentrating hormone neurons in experimental sleeping sickness. Neuroscience 2015; 290:185-95. [PMID: 25595977 DOI: 10.1016/j.neuroscience.2014.12.066] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/12/2014] [Accepted: 12/23/2014] [Indexed: 01/24/2023]
Abstract
Human African trypanosomiasis or sleeping sickness is a severe, neglected tropical disease caused by the extracellular parasite Trypanosoma brucei. The disease, which leads to chronic neuroinflammation, is characterized by sleep and wake disturbances, documented also in rodent models. In rats and mice infected with Trypanosoma brucei brucei, we here tested the hypothesis that the disease could target neurons of the lateral hypothalamus (LH) containing orexin (OX)-A or melanin-concentrating hormone (MCH), implicated in sleep/wake regulation. In the cerebrospinal fluid of infected rats, the OX-A level was significantly decreased early after parasite neuroinvasion, and returned to the control level at an advanced disease stage. The number of immunohistochemically characterized OX-A and MCH neurons decreased significantly in infected rats during disease progression and in infected mice at an advanced disease stage. A marked reduction of the complexity of dendritic arborizations of OX-A neurons was documented in infected mice. The evaluation of NeuN-immunoreactive neurons did not reveal significant neuronal loss in the LH of infected mice, thus suggesting a potential selective vulnerability of OX-A and MCH neurons. Immunophenotyping and quantitative analysis showed in infected mice marked activation of microglial cells surrounding OX-A neurons. Day/night oscillation of c-Fos baseline expression was used as marker of OX-A neuron activity in mice. In control animals Fos was expressed in a higher proportion of OX-A neurons in the night (activity) phase than in the day (rest) phase. Interestingly, in infected mice the diurnal spontaneous Fos oscillation was reversed, with a proportion of OX-A/Fos neurons significantly higher at daytime than at nighttime. Altogether the findings reveal a progressive decrease of OX-A and MCH neurons and dysregulation of OX-A neuron diurnal activity in rodent models of sleeping sickness. The data point to the involvement of these peptidergic neurons in the pathogenesis of sleep/wake alterations in the disease and to their vulnerability to inflammatory signaling.
Collapse
Affiliation(s)
- M Palomba
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - P F Seke-Etet
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - C Laperchia
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - L Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Y-Z Xu
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - V Colavito
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - G Grassi-Zucconi
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - M Bentivoglio
- Department of Neurological and Movement Sciences, University of Verona, Italy.
| |
Collapse
|
39
|
Nobunaga M, Obukuro K, Kurauchi Y, Hisatsune A, Seki T, Tsutsui M, Katsuki H. High fat diet induces specific pathological changes in hypothalamic orexin neurons in mice. Neurochem Int 2014; 78:61-6. [PMID: 25195718 DOI: 10.1016/j.neuint.2014.09.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/05/2014] [Accepted: 09/01/2014] [Indexed: 11/17/2022]
Abstract
Loss of orexin neurons in the hypothalamus is a prominent feature of narcolepsy and several other neurological conditions. We have recently demonstrated that sleep deprivation stimulates local nitric oxide (NO) production by neuronal NO synthase in the lateral hypothalamus, which leads to selective degeneration of orexin neurons accompanied by formation of orexin-immunoreactive aggregates. Here we analyzed whether lifestyle-related conditions other than sleep deprivation could trigger similar pathological changes in orexin neurons. Four-week-old male C57BL/6 mice were fed with high fat diet (HFD) for 8 weeks. Immunohistochemical analysis revealed that the number of orexin-immunopositive neurons was significantly decreased by HFD intake, whereas the number of melanin-concentrating hormone-immunopositive neurons was unchanged. In addition, HFD promoted formation of intracellular orexin-immunoreactive aggregates in a subset of orexin neurons. We also confirmed that expression of inducible NO synthase (iNOS) in the hypothalamus was upregulated in response to HFD intake. Notably, loss of orexin-immunopositive neurons and formation of orexin-immunoreactive aggregates were not observed in iNOS knockout mice fed with HFD. These results indicate that inappropriate dietary conditions could trigger specific neuropathological events in orexin neurons in an iNOS-dependent manner.
Collapse
Affiliation(s)
- Mizuki Nobunaga
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Kanae Obukuro
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yuki Kurauchi
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto 860-8555, Japan; Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program", Kumamoto University, Kumamoto 862-0973, Japan
| | - Akinori Hisatsune
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto 860-8555, Japan; Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program", Kumamoto University, Kumamoto 862-0973, Japan
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan.
| |
Collapse
|
40
|
Kostin A, McGinty D, Szymusiak R, Alam MN. Sleep-wake and diurnal modulation of nitric oxide in the perifornical-lateral hypothalamic area: real-time detection in freely behaving rats. Neuroscience 2013; 254:275-84. [PMID: 24056193 DOI: 10.1016/j.neuroscience.2013.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) has been implicated in the regulation of sleep. The perifornical-lateral hypothalamic area (PF-LHA) is a key wake-promoting region and contains neurons that are active during behavioral or cortical activation. Recently, we found higher levels of NO metabolites (NOx), an indirect measure of NO levels, in the PF-LHA during prolonged waking (SD). However, NO is highly reactive and diffuses rapidly and the NOx assay is not sensitive enough to detect rapid-changes in NO levels across spontaneous sleep-waking states. We used a novel Nafion®-modified Platinum (NF-PT) electrode for real-time detection of NO levels in the PF-LHA across sleep-wake cycles, dark-light phases, and during SD. Sprague-Dawley male rats were surgically prepared for chronic sleep-wake recording and implantation of NF-PT electrode into the PF-LHA. Electroencephalogram (EEG), electromyogram (EMG), and electrochemical current generated by NF-PT electrode were continuously acquired for 5-7days including one day with 3h of SD. In the PF-LHA, NO levels exhibited a waking>rapid eye movement (REM)>non-rapid eye movement (nonREM) sleep pattern (0.56±0.03μM>0.47±0.02μM>0.42±0.02μM; p<0.01). NO levels were also higher during the dark- as compared to the light-phase (0.53±0.03μM vs. 0.44±0.02μM; p<0.01). NO levels increased during 3h of SD as compared to undisturbed control (0.58±0.04μM vs. 0.47±0.01μM; p<0.05). The findings indicate that in the PF-LHA, NO is produced during behavioral or cortical activation. Since elevated levels of NO inhibits most of the PF-LHA neurons that are active during cortical activation, these findings support a hypothesis that NO produced in conjunction with the activation of PF-LHA neurons during waking/SD, inhibits the same neuronal population to promote sleep.
Collapse
Affiliation(s)
- A Kostin
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA 91343, USA
| | | | | | | |
Collapse
|