1
|
Lozano-Tovar S, Cremascoli R, Nuccetelli M, Sancesario G, Cattaldo S, Prina E, Verde F, Cappelli S, Bernardini S, Mercuri NB, Liguori C. Cerebrospinal-fluid Orexin-A levels in different neurocognitive disorders: a comparison study. Neurol Sci 2025:10.1007/s10072-025-08148-0. [PMID: 40198471 DOI: 10.1007/s10072-025-08148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/23/2025] [Indexed: 04/10/2025]
Abstract
In the present study, we investigated the differences in cerebrospinal fluid (CSF) orexin-A levels among patients with different neurocognitive disorders, such as mild or moderate to severe Alzheimer's disease (AD; mAD, msAD, respectively), behavioral variants of frontotemporal dementia (bv-FTD), non-fluent primary aphasia (NFPA), and idiopathic normal pressure hydrocephalus (iNPH). A total of 214 participants were evaluated (mAD, 45; msAD, 31; bv-FTD, 12; NFPA, 22; iNPH, 13; non-demented elderly controls, 91). The highest CSF orexin-A levels were found in iNPH patients (263.31 ± 56.89 pg/mL). Patients affected by NFPA (210.86 ± 61.99 pg/mL), iNPH, and msAD (173.04 ± 19.76 pg/mL) showed higher CSF orexin-A concentrations than controls (145.18 ± 27.01pg/mL) (p < 0.001). Bv-FTD (190.12 ± 100.84 pg/mL) and mAD (130.76 ± 21.70 pg/mL) patients showed no significant differences in CSF orexin-A levels compared with controls. mAD patients showed also lower CSF orexin-A concentrations than all other patient groups.In conclusion, orexin-A presents different CSF levels among neurocognitive disorders. The mechanisms underlying this difference vary and may include sleep-wake cycle impairment, behavioral disturbances, and CSF dynamics. The development of drugs that antagonize the orexin system could open a new frontier of research linking orexin neurotransmission to neurocognitive disorders.
Collapse
Affiliation(s)
- Susana Lozano-Tovar
- Facultad de Psicología, Universidad Nacional Autónoma de México (UNAM), Circuito Ciudad Universitaria Avenida, C.U, Mexico City, 04510, Mexico
| | - Riccardo Cremascoli
- Unit of Neurology and Neurorehabilitation, IRCCS, Istituto Auxologico Italiano, San Giuseppe Hospital, 28824, Verbania, Italy
| | - Marzia Nuccetelli
- Department of Clinical Biochemistry and Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Sancesario
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Stefania Cattaldo
- Laboratory of Clinical Neurobiology, IRCCS, Istituto Auxologico Italiano, San Giuseppe Hospital, 28824, Verbania, Italy
| | - Elisa Prina
- Department of Neurology and Laboratory of Neuroscience, IRCCS, Istituto Auxologico Italiano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, IRCCS, Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Simone Cappelli
- Laboratory of Psychology, IRCCS, Istituto Auxologico Italiano, San Giuseppe Hospital, 28824, Verbania, Italy
| | - Sergio Bernardini
- Unit of Neurology and Neurorehabilitation, IRCCS, Istituto Auxologico Italiano, San Giuseppe Hospital, 28824, Verbania, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy.
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Viale Oxford 81, 00133, Rome, Italy.
| |
Collapse
|
2
|
Zhang J, Jin K, Chen B, Cheng S, Jin J, Yang X, Lu J, Song Q. Sex-dimorphic functions of orexin in neuropsychiatric disorders. Heliyon 2024; 10:e36402. [PMID: 39253145 PMCID: PMC11382083 DOI: 10.1016/j.heliyon.2024.e36402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
The orexin system regulates a variety of physiological functions, including the sleep-wake cycle, addiction, foraging behavior, stress and cognitive functioning. Orexin levels in central and peripheral are related to the pathogenesis of many diseases, most notably the narcolepsy, eating disorders, stress-related psychiatric disorders, and neurodegenerative diseases. Recently, it has been reported that the orexin system is distinctly sexually dimorphic, and is strongly associated with neuropsychiatric disorders. In this review, we analyzed advancements in the sex differences in the orexin system and their connection to psychoneurological conditions. Considering the scarcity of research in this domain, more research is imperative to reveal the underlying mechanisms.
Collapse
Affiliation(s)
- Jinghan Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Kangyu Jin
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
| | - Bing Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
| | - Shangping Cheng
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
| | - Jinfan Jin
- Department of Psychiatry, Lishui Second People's Hospital, Lishui, Zhejiang, 323000, China
| | - Xiaolan Yang
- Department of Psychiatry, Lishui Second People's Hospital, Lishui, Zhejiang, 323000, China
| | - Jing Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China
| | - Qinghai Song
- Department of Psychiatry, Lishui Second People's Hospital, Lishui, Zhejiang, 323000, China
| |
Collapse
|
3
|
Bohid S, Ali LK, Romero-Leguizamón CR, Langkilde AE, Dos Santos AB, Kohlmeier KA. Sex-dependent effects of monomeric α-synuclein on calcium and cell death of lateral hypothalamic mouse neurons are altered by orexin. Mol Cell Neurosci 2024; 129:103934. [PMID: 38701995 DOI: 10.1016/j.mcn.2024.103934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/25/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024] Open
Abstract
Parkinson's Disease (PD) patients experience sleeping disorders in addition to the disease-defining symptomology of movement dysfunctions. The prevalence of PD is sex-based and presence of sleeping disorders in PD also shows sex bias with a stronger phenotype in males. In addition to loss of dopamine-containing neurons in the striatum, arousal-related, orexin-containing neurons in the lateral hypothalamus (LH) are lost in PD, which could contribute to state-related disorders. As orexin has been shown to be involved in sleeping disorders and to have neuroprotective effects, we asked whether orexin could protect sleep-related LH neurons from damage putatively from the protein α-synuclein (α-syn), which is found at high levels in the PD brain and that we have shown is associated with putatively excitotoxic rises in intracellular calcium in brainstem sleep-controlling nuclei, especially in males. Accordingly, we monitored intracellular calcium transients induced by α-syn and whether concurrent exposure to orexin affected those transients in LH cells of the mouse brain slice using calcium imaging. Further, we used an assay of cell death to determine whether LH cell viability was influenced when α-syn and orexin were co-applied when compared to exposure to α-syn alone. We found that excitatory calcium events induced by α-syn were reduced in amplitude and frequency when orexin was co-applied, and when data were evaluated by sex, this effect was found to be greater in females. In addition, α-syn exposure was associated with cell death that was higher in males, and interestingly, reduced cell death was noted when orexin was present, which did not show a sex bias. We interpret our findings to indicate that orexin is protective to α-syn-mediated damage to hypothalamic neurons, and the actions of orexin on α-syn-induced cellular effects differ between sexes, which could underlie sex-based differences in sleeping disorders in PD.
Collapse
Affiliation(s)
- Sara Bohid
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Lara Kamal Ali
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Cesar Ramon Romero-Leguizamón
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Annette E Langkilde
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Altair Brito Dos Santos
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark.
| |
Collapse
|
4
|
Gao Y, Zhou Q, Li H, Zhao Y, Zhu H, Zhang X, Li Y. Melanin-Concentrating Hormone Is Associated With Delayed Neurocognitive Recovery in Older Adult Patients With Preoperative Sleep Disorders Undergoing Spinal Anesthesia. Anesth Analg 2024; 138:579-588. [PMID: 38051670 DOI: 10.1213/ane.0000000000006768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
BACKGROUND Aging and preoperative sleep disorders are the main risk factors affecting postoperative cognitive outcomes. However, the pathogenesis of delayed neurocognitive recovery after surgery remains ambiguous, and there is still a lack of potential biomarkers for delayed neurocognitive recovery in older adult patients with preoperative sleep disorders. Our study aimed to explore the relationship between melanin-concentrating hormone (MCH) and delayed neurocognitive recovery early after surgery in older adult patients with preoperative sleep disorders. METHODS In this monocentric prospective observational study, 156 older adult patients (aged 65 years or older) with preoperative sleep disorders undergoing elective total hip arthroplasty (THA) or total knee arthroplasty (TKA) were included at an academic medical center in Inner Mongolia, China, from October 2021 to November 2022, and all patients underwent spinal anesthesia. The Pittsburgh Sleep Quality Index (PSQI) was applied to assess the preoperative sleep quality of all patients, and preoperative sleep disorders were defined as a score of PSQI >5. We measured the levels of cerebrospinal fluid (CSF) MCH and plasma MCH of all patients. The primary outcome was delayed neurocognitive recovery early after surgery. All patients received cognitive function assessment through the Montreal Cognitive Assessment (MoCA) 1 day before and 7 days after surgery (postoperative day 7 [POD7]). Delayed neurocognitive recovery was defined as a score of POD7 MoCA <26. The potential confounders included variables with P < .2 in the univariate logistic analysis, as well as the important risk factors of delayed neurocognitive recovery reported in the literature. Multivariable logistic regression model based on the Enter method assessed the association of MCH and delayed neurocognitive recovery in older adult patients with preoperative sleep disorders. RESULTS Fifty-nine (37.8%) older adult patients with preoperative sleep disorders experienced delayed neurocognitive recovery at POD7. Increase in CSF MCH levels (odds ratio [OR] for an increase of 1 pg/mL = 1.16, 95% confidence interval [CI], 1.09-1.23, P < .001) and decrease in plasma MCH levels (OR for an increase of 1 pg/mL = 0.92, 95% CI, 0.86-0.98, P = .003) were associated with delayed neurocognitive recovery, after adjusting for age, sex, education, baseline MoCA scores, American Society of Anesthesiologists (ASA) grade, and coronary heart disease (CHD). CONCLUSIONS In older adult patients with preoperative sleep disorders, MCH is associated with the occurrence of delayed neurocognitive recovery after surgery. Preoperative testing of CSF MCH or plasma MCH may increase the likelihood of identifying the high-risk population for delayed neurocognitive recovery in older adult patients with preoperative sleep disorders.
Collapse
Affiliation(s)
- Yi Gao
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, China
| | - Qi Zhou
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, China
| | - Haibo Li
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, China
| | - Yunjiao Zhao
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Hongyan Zhu
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, China
| | - Xizhe Zhang
- From the Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, China
| | - Yun Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin Research Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
5
|
Johnson CE, Duncan MJ, Murphy MP. Sex and Sleep Disruption as Contributing Factors in Alzheimer's Disease. J Alzheimers Dis 2024; 97:31-74. [PMID: 38007653 PMCID: PMC10842753 DOI: 10.3233/jad-230527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease (AD) affects more women than men, with women throughout the menopausal transition potentially being the most under researched and at-risk group. Sleep disruptions, which are an established risk factor for AD, increase in prevalence with normal aging and are exacerbated in women during menopause. Sex differences showing more disrupted sleep patterns and increased AD pathology in women and female animal models have been established in literature, with much emphasis placed on loss of circulating gonadal hormones with age. Interestingly, increases in gonadotropins such as follicle stimulating hormone are emerging to be a major contributor to AD pathogenesis and may also play a role in sleep disruption, perhaps in combination with other lesser studied hormones. Several sleep influencing regions of the brain appear to be affected early in AD progression and some may exhibit sexual dimorphisms that may contribute to increased sleep disruptions in women with age. Additionally, some of the most common sleep disorders, as well as multiple health conditions that impair sleep quality, are more prevalent and more severe in women. These conditions are often comorbid with AD and have bi-directional relationships that contribute synergistically to cognitive decline and neuropathology. The association during aging of increased sleep disruption and sleep disorders, dramatic hormonal changes during and after menopause, and increased AD pathology may be interacting and contributing factors that lead to the increased number of women living with AD.
Collapse
Affiliation(s)
- Carrie E. Johnson
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
| | - Marilyn J. Duncan
- University of Kentucky, College of Medicine, Department of Neuroscience, Lexington, KY, USA
| | - M. Paul Murphy
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY, USA
| |
Collapse
|
6
|
Kim LJ, Pho H, Anokye-Danso F, Ahima RS, Pham LV, Polotsky VY. The effect of diet-induced obesity on sleep and breathing in female mice. Sleep 2023; 46:zsad158. [PMID: 37262435 PMCID: PMC10424169 DOI: 10.1093/sleep/zsad158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/24/2023] [Indexed: 06/03/2023] Open
Abstract
Obesity and male sex are main risk factors for sleep-disordered breathing (SDB). We have shown that male diet-induced obesity (DIO) mice develop hypoventilation, sleep apnea, and sleep fragmentation. The effects of DIO on breathing and sleep architecture in females have not been investigated. We hypothesized that female mice are less susceptible to the detrimental effects of DIO on sleep and SDB compared to males. Female DIO-C57BL/6J and lean C57BL/6J mice underwent 24-hour metabolic studies and were exposed to 8% CO2 to measure the hypercapnic ventilatory response (HCVR), and sleep studies. Ventilatory response to arousals was calculated as ratio of the average and peak minute ventilation (VE) during each arousal relative to the baseline VE. Breathing stability was measured with Poincaré plots of VE. Female obesity was associated with decreased metabolism, indicated by reduced oxygen consumption (VO2) and CO2 production (VCO2). VE in 8% CO2 and HCVR were significantly attenuated during wakefulness. NREM sleep duration was reduced in DIO mice, but REM sleep was preserved. Ventilation during NREM and REM sleep was augmented compared to lean mice. Arousal frequency was similar between groups. Obesity increased the frequency of spontaneous arousals, whereas the apnea index was 4-fold reduced in DIO compared to lean mice. Obesity decreased pre- and post-apnea arousals. Obese mice had more stable breathing with reduced ventilatory response to arousals, compared to lean females. We conclude that obese female mice are protected against SDB, which appears to be related to an attenuated CO2 responsiveness, compared to the lean state.
Collapse
Affiliation(s)
- Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| |
Collapse
|
7
|
Ten-Blanco M, Flores Á, Cristino L, Pereda-Pérez I, Berrendero F. Targeting the orexin/hypocretin system for the treatment of neuropsychiatric and neurodegenerative diseases: from animal to clinical studies. Front Neuroendocrinol 2023; 69:101066. [PMID: 37015302 DOI: 10.1016/j.yfrne.2023.101066] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/06/2023]
Abstract
Orexins (also known as hypocretins) are neuropeptides located exclusively in hypothalamic neurons that have extensive projections throughout the central nervous system and bind two different G protein-coupled receptors (OX1R and OX2R). Since its discovery in 1998, the orexin system has gained the interest of the scientific community as a potential therapeutic target for the treatment of different pathological conditions. Considering previous basic science research, a dual orexin receptor antagonist, suvorexant, was the first orexin agent to be approved by the US Food and Drug Administration to treat insomnia. In this review, we discuss and update the main preclinical and human studies involving the orexin system with several psychiatric and neurodegenerative diseases. This system constitutes a nice example of how basic scientific research driven by curiosity can be the best route to the generation of new and powerful pharmacological treatments.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Neurosciences Institute, University of Barcelona and Bellvitge University Hospital-IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Inmaculada Pereda-Pérez
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Berrendero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
8
|
Ghani U. Azole inhibitors of mushroom and human tyrosinases: Current advances and prospects of drug development for melanogenic dermatological disorders. Eur J Med Chem 2022; 239:114525. [PMID: 35717871 DOI: 10.1016/j.ejmech.2022.114525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022]
Abstract
Azoles are a famous and promising class of drugs for treatment of a range of ailments especially fungal infections. A wide variety of azole derivatives are also known to exhibit tyrosinase inhibition, some of which possess promising activity with potential for treatment of dermatological disorders such as post-inflammatory hyperpigmentation, nevus, flecks, melasma, and melanoma. Recently, thiazolyl-resorcinol derivatives have demonstrated potent human tyrosinase inhibition with a safe and effective therapeutic profile for treatment of skin hyperpigmentation in humans, which are currently under clinical trials. If approved these derivatives would be the first azole drugs to be used for treatment of skin hyperpigmentation. Although the scientific literature has been witnessing general reviews on tyrosinase inhibitors to date, there is none that specifically and comprehensively discusses azole inhibitors of tyrosinase. Appreciating such potential of azoles, this focused review highlights a wide range of their derivatives with promising mushroom and human tyrosinase inhibitory activities and clinical potential for treatment of melanogenic dermatological disorders. Presently, these disorders have been treated with kojic acid, hydroquinone and other drugs, the design and development of which are based on their ability to inhibit mushroom tyrosinase. The active sites of mushroom and human tyrosinases carry structural differences which affect substrate or inhibitor binding. For this reason, kojic acid and other drugs pose efficacy and safety issues since they were originally developed using mushroom tyrosinase and have been clinically used on human tyrosinase. Design and development of tyrosinase inhibitors should be based on human tyrosinase, however, there are challenges in obtaining the human enzyme and understanding its structure and function. The review discusses these challenges that encompass structural and functional differences between mushroom and human tyrosinases and the manner in which they are inhibited. The review also gauges promising azole derivatives with potential for development of drugs against skin hyperpigmentation by analyzing and comparing their tyrosinase inhibitory activities against mushroom and human tyrosinases, computational data, and clinical profile where available. It aims to lay groundwork for development of new azole drugs for treatment of skin hyperpigmentation, melanoma, and related dermatological disorders.
Collapse
Affiliation(s)
- Usman Ghani
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh, 12372, Saudi Arabia.
| |
Collapse
|
9
|
Electroacupuncture Enhances Neuroplasticity by Regulating the Orexin A-Mediated cAMP/PKA/CREB Signaling Pathway in Senescence-Accelerated Mouse Prone 8 (SAMP8) Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8694462. [PMID: 35154573 PMCID: PMC8837456 DOI: 10.1155/2022/8694462] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/03/2022] [Indexed: 11/18/2022]
Abstract
Learning and memory disorders and decreased neuroplasticity are the main clinical manifestations of age-induced cognitive dysfunction. Orexin A (OxA) has been reported to show abnormally elevated expression in the cerebrospinal fluid (CSF) of patients with Alzheimer's disease (AD) and to be associated with cognitive impairment. Here, we further assessed whether the excitatory neurotransmitter OxA is involved in neuroplasticity and cognitive function in senescence-accelerated mouse prone 8 (SAMP8) mice. In this study, we investigated the mechanism of OxA by using behavioral tests, CSF microdialysis, immunofluorescence, toluidine blue staining, gene silencing, transmission electron microscopy, and Western blotting. The results showed that 10 Hz electroacupuncture (EA) effectively alleviated learning and memory impairment in 7-month-old SAMP8 mice, reduced OxA levels in the CSF, increased the level of the neurotransmitter glutamate, alleviated pathological damage to hippocampal tissue, improved the synaptic structure, enhanced synaptic transmission, and regulated the expression of cAMP/PKA/CREB signaling pathway-related proteins. These results suggest that EA enhances neuroplasticity in SAMP8 mice by regulating the OxA-mediated cAMP/PKA/CREB signaling pathway, thus improving cognitive function. These findings suggest that EA may be beneficial for the prevention and treatment of age-induced cognitive impairment.
Collapse
|
10
|
Matzeu A, Martin-Fardon R. Understanding the Role of Orexin Neuropeptides in Drug Addiction: Preclinical Studies and Translational Value. Front Behav Neurosci 2022; 15:787595. [PMID: 35126069 PMCID: PMC8811192 DOI: 10.3389/fnbeh.2021.787595] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/29/2021] [Indexed: 12/22/2022] Open
Abstract
Orexins (also known as hypocretins) are neuropeptides that participate in the regulation of energy metabolism, homeostasis, sleep, feeding, stress responses, arousal, and reward. Particularly relevant to the scope of the present review is the involvement of the orexin system in brain mechanisms that regulate motivation, especially highly motivated behavior, arousal, and stress, making it an ideal target for studying addiction and discovering treatments. Drug abuse and misuse are thought to induce maladaptive changes in the orexin system, and these changes might promote and maintain uncontrolled drug intake and contribute to relapse. Dysfunctional changes in this neuropeptidergic system that are caused by drug use might also be responsible for alterations of feeding behavior and the sleep-wake cycle that are commonly disrupted in subjects with substance use disorder. Drug addiction has often been associated with an increase in activity of the orexin system, suggesting that orexin receptor antagonists may be a promising pharmacological treatment for substance use disorder. Substantial evidence has shown that single orexin receptor antagonists that are specific to either orexin receptor 1 or 2 can be beneficial against drug intake and relapse. Interest in the efficacy of dual orexin receptor antagonists, which were primarily developed to treat insomnia, has grown in the field of drug addiction. Treatments that target the orexin system may be a promising strategy to reduce drug intake, mitigate relapse vulnerability, and restore “normal” physiological functions, including feeding and sleep. The present review discusses preclinical and clinical evidence of the involvement of orexins in drug addiction and possible beneficial pharmacotherapeutic effects of orexin receptor antagonists to treat substance use disorder.
Collapse
|
11
|
Guo L, Hu A, Zhao X, Xiang X. Reduction of Orexin-A Is Associated With Anxiety and the Level of Depression of Male Methamphetamine Users During the Initial Withdrawal Period. Front Psychiatry 2022; 13:900135. [PMID: 35859609 PMCID: PMC9289462 DOI: 10.3389/fpsyt.2022.900135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Orexin has been linked to the regulation of reward and motivation in animals, but there have been few human studies to validate its regulatory effects. We aimed to determine how orexin-A levels changed during different stages of withdrawal, as well as the relationship between orexin-A levels and withdrawal symptoms in male METH users. METHODS This study included 76 METH users and 35 control participants. The METH users were divided into three groups: group 1 (abstinence within 1 week, n = 23), group 2 (abstinence between 1 week and 3 months, n = 38), and group 3 (abstinence over 3 months, n = 15). At baseline and 1 month of enrollment, the plasma orexin-A level was examined. To track the withdrawal symptoms, self-report questionnaires (anxiety, depression, craving, and sleep quality) were collected at two points. RESULTS The orexin-A levels of groups 1 (p < 0.001) and 2 (p < 0.001) were lower than that of the controls at baseline but not group 3. One month later, the orexin-A levels of group 2 (p < 0.05) significantly increased, while no significant changes in those of groups 1 and 3 were observed. Furthermore, the orexin-A levels of group 1 were positively linked with depression (p < 0.01) and anxiety (p < 0.01) at baseline. CONCLUSIONS The decrease in orexin-A levels was only transitory during the initial abstinence phase, and it was eventually restored near to normal with continued abstinence among the male METH users. Furthermore, a lower concentration of orexin-A may serve as a risk factor for negative emotions during METH withdrawal.
Collapse
Affiliation(s)
- Lei Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Aqian Hu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoxi Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaojun Xiang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
12
|
Mehramiz M, Porter T, Laws SM, Rainey-Smith SR. Sleep, Sirtuin 1 and Alzheimer's disease: A review. AGING BRAIN 2022; 2:100050. [PMID: 36908890 PMCID: PMC9997138 DOI: 10.1016/j.nbas.2022.100050] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Sleep plays a major role in brain health, and cognition. Disrupted sleep is a well-described symptom of Alzheimer's disease (AD). However, accumulating evidence suggests suboptimal sleep also increases AD risk. The deacetylase Sirtuin 1 (Sirt 1), encoded by the SIRT1 gene, impacts sleep via its relationship to wake-sleep neurotransmitters and somnogens. Evidence from animal and human studies supports a significant and complex relationship between sleep, Sirt 1/ SIRT1 and AD. Numerous hypotheses attempt to explain the critical impact of Sirt 1/ SIRT1 on wake- and sleep- promoting neurons, their related mechanisms and neurotransmitters. However, there is a paucity of studies assessing the interaction between sleep and Sirt 1/ SIRT1, as a principal component of sleep regulation, on AD pathology. In this review, we explore the potential association between Sirt 1/ SIRT1, sleep, and AD aetiology. Given sleep is a likely modifiable risk factor for AD, and recent studies suggest Sirt 1/ SIRT1 activation can be modulated by lifestyle or dietary approaches, further research in this area is required to explore its potential as a target for AD prevention and treatment.
Collapse
Affiliation(s)
- Mehrane Mehramiz
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
13
|
Cerebrospinal fluid orexin in Alzheimer's disease: a systematic review and meta-analysis. Sleep Med 2021; 85:230-238. [PMID: 34364094 DOI: 10.1016/j.sleep.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE/BACKGROUND A growing body of evidence suggests that sleep and Alzheimer's disease (AD) have a bi-directional relationship. Emerging research also suggests that orexin, a key neurotransmitter involved in sleep-wake regulation, may be altered in persons with AD, however results have not been consistent across prior studies. This investigation was conducted to both evaluate the aggregate literature to minimize the risk of bias and identify potential factors associated with heterogeneity across studies. METHODS Systematic review identified relevant investigations that compared cerebrospinal fluid orexin in persons with AD and controls. Meta-analysis (random effects model) compared effect size (Hedge's g) for orexin between AD and controls. Meta-regression was additionally performed for key variables of interest to evaluate potential causes of heterogeneity among studies. RESULTS 17 studies were identified that met inclusion/exclusion criteria. Evidence of publication bias was not identified. Non-significant increases in orexin were observed in AD relative to controls, with moderate to large heterogeneity among studies (Hedge's g = 0.20, p = 0.136, I2 = 72.6%). Meta-regression demonstrated both year of publication (β = 0.055, p = 0.020) and effect size for phosphorylated tau in AD versus controls (β = 0.417, p = 0.031) were associated with differences in orexin. CONCLUSIONS Results do not support broad differences in orexin in AD compared to controls, however, evolving diagnostic criteria may have affected findings across studies. Future research that examines orexin in AD over the longitudinal course of the disorder and explores potential links between phosphorylated tau and orexin are indicated.
Collapse
|
14
|
Lonstein JS, Linning-Duffy K, Tang Y, Moody A, Yan L. Impact of daytime light intensity on the central orexin (hypocretin) system of a diurnal rodent (Arvicanthis niloticus). Eur J Neurosci 2021; 54:4167-4181. [PMID: 33899987 DOI: 10.1111/ejn.15248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 11/29/2022]
Abstract
The neuropeptide orexin/hypocretin is implicated in sleep and arousal, energy expenditure, reward, affective state and cognition. Our previous work using diurnal Nile grass rats (Arvicanthis niloticus) found that orexin mediates the effects of environmental light, particularly daytime light intensity, on affective and cognitive behaviours. The present study further investigated how daytime light intensity affects the central orexin system in male and female grass rats. Subjects were housed for 4 weeks in 12:12 hr dim light:dark (50 lux, dimLD) or in 12:12 hr bright light:dark cycle (1000 lux, brightLD). Day/night fluctuations in some orexin measures were also assessed. Despite similar hypothalamic prepro-orexin mRNA expression across all conditions, there were significantly more orexin-immunoreactive neurons, larger somata, greater optical density or higher orexin A content at night (ZT14) than during the day (ZT2), and/or in animals housed in brightLD compared to dimLD. Grass rats in brightLD also had higher cisternal CSF levels of orexin A. Furthermore, orexin receptor OX1R and OX2R proteins in the medial prefrontal cortex were higher in brightLD than dimLD males, but lower in brightLD than dimLD females. In the CA1 and dorsal raphe nucleus, females had higher OX1R than males without any significant effects of light condition, and OX2R levels were unaffected by sex or light. These results reveal that daytime light intensity alters the central orexin system of both male and female diurnal grass rats, sometimes sex-specifically, and provides insight into the mechanisms underlying how daytime light intensity impacts orexin-regulated functions.
Collapse
Affiliation(s)
- Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Yuping Tang
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anna Moody
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
15
|
Ruiz-Viroga V, Urbanavicius J, Torterolo P, Lagos P. In vivo uptake of a fluorescent conjugate of melanin-concentrating hormone in the rat brain. J Chem Neuroanat 2021; 114:101959. [PMID: 33848617 DOI: 10.1016/j.jchemneu.2021.101959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 04/03/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide synthesized by posterior hypothalamic and incerto-hypothalamic neurons that project throughout the central nervous system. The MCHergic system modulates several important functions such as feeding behavior, mood and sleep. MCH exerts its biological functions through interaction with the MCHR-1 receptor, the only functional receptor present in rodents. The internalization process of MCHR-1 triggered by MCH binding was described in vitro in non-neuronal heterologous systems with over-expression of MCHR-1. Reports of in vivo MCHR-1 internalization dynamics are scarce, however, this is an important process to explore based on the critical functions of the MCHergic system. We had previously determined that 60 min after intracerebroventricular (i.c.v.) microinjections of MCH conjugated with fluorophore rhodamine (R-MCH), the dorsal and median raphe nucleus presented R-MCH positive labeled neurons. In the present work, we further studied the in vivo uptake process focusing on the distribution and time-dependent pattern of R-MCH positive cells 10, 20 and 60 min (T10, T20 and T60, respectively) after i.c.v. microinjection of R-MCH. We also explored this uptake process to see whether it was receptor- and clathrin-dependent and examined the phenotype of R-MCH positive cells and their proximity to MCHergic fibers. We found a great number of R-MCH positive cells with high fluorescence intensity in the lateral septum, nucleus accumbens and hippocampus at T20 and T60 (but not at T10), while a lower number with low intensity was observed in the dorsal raphe nucleus. At T20, in rats pre-treated with a MCHR-1 antagonist (ATC-0175) or with phenylarsine oxide (PAO), a clathrin endocytosis inhibitor, a robust decrease (> 50 %) of R-MCH uptake occurred in these structures. The R-MCH positive cells were identified as neurons (NeuN positive, GFAP negative) and some MCHergic fibers run in the vicinities of them. We concluded that neurons localized at structures that were close to the ventricular surfaces could uptake R-MCH in vivo through a receptor-dependent and clathrin-mediated process. Our results support volume transmission of MCH through the cerebrospinal fluid to reach distant targets. Finally, we propose that R-MCH would be an effective tool to study MCH-uptake in vivo.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jessika Urbanavicius
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
16
|
Van Drunen R, Eckel-Mahan K. Circadian Rhythms of the Hypothalamus: From Function to Physiology. Clocks Sleep 2021; 3:189-226. [PMID: 33668705 PMCID: PMC7931002 DOI: 10.3390/clockssleep3010012] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
The nearly ubiquitous expression of endogenous 24 h oscillations known as circadian rhythms regulate the timing of physiological functions in the body. These intrinsic rhythms are sensitive to external cues, known as zeitgebers, which entrain the internal biological processes to the daily environmental changes in light, temperature, and food availability. Light directly entrains the master clock, the suprachiasmatic nucleus (SCN) which lies in the hypothalamus of the brain and is responsible for synchronizing internal rhythms. However, recent evidence underscores the importance of other hypothalamic nuclei in regulating several essential rhythmic biological functions. These extra-SCN hypothalamic nuclei also express circadian rhythms, suggesting distinct regions that oscillate either semi-autonomously or independent of SCN innervation. Concurrently, the extra-SCN hypothalamic nuclei are also sensitized to fluctuations in nutrient and hormonal signals. Thus, food intake acts as another powerful entrainer for the hypothalamic oscillators' mediation of energy homeostasis. Ablation studies and genetic mouse models with perturbed extra-SCN hypothalamic nuclei function reveal their critical downstream involvement in an array of functions including metabolism, thermogenesis, food consumption, thirst, mood and sleep. Large epidemiological studies of individuals whose internal circadian cycle is chronically disrupted reveal that disruption of our internal clock is associated with an increased risk of obesity and several neurological diseases and disorders. In this review, we discuss the profound role of the extra-SCN hypothalamic nuclei in rhythmically regulating and coordinating body wide functions.
Collapse
Affiliation(s)
- Rachel Van Drunen
- MD Anderson UTHealth School Graduate School of Biomedical Sciences, Houston TX 77030, USA;
- Brown Foundation Institute of Molecular Medicine University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Kristin Eckel-Mahan
- MD Anderson UTHealth School Graduate School of Biomedical Sciences, Houston TX 77030, USA;
- Brown Foundation Institute of Molecular Medicine University of Texas McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
17
|
Kent BA, Feldman HH, Nygaard HB. Sleep and its regulation: An emerging pathogenic and treatment frontier in Alzheimer's disease. Prog Neurobiol 2021; 197:101902. [PMID: 32877742 PMCID: PMC7855222 DOI: 10.1016/j.pneurobio.2020.101902] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/19/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
A majority of patients with Alzheimer's disease (AD) experience some form of sleep disruption, including nocturnal sleep fragmentation, increased daytime napping, decreased slow-wave sleep (SWS, stage N3), and decreased rapid-eye-movement sleep (REM). Clinical studies are investigating whether such sleep disturbances are a consequence of the underlying disease, and whether they also contribute to the clinical and pathological manifestations of AD. Emerging research has provided a direct link between several of these sleep disruptions and AD pathophysiology, suggesting that treating sleep disorders in this population may target basic mechanisms of the disease. Here, we provide a comprehensive review of sleep disturbances associated with the spectrum of AD, ranging from the preclinical stages through dementia. We discuss how sleep interacts with AD pathophysiology and, critically, whether sleep impairments can be targeted to modify the disease course in a subgroup of affected AD patients. Ultimately, larger studies that fully utilize new diagnostic and experimental tools will be required to better define the most relevant sleep disturbance to target in AD, the interventions that best modulate this target symptom, and whether successful early intervention can modify AD risk and prevent dementia.
Collapse
Affiliation(s)
- Brianne A Kent
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Division of Neurology, University of British Columbia, Vancouver, Canada.
| | - Howard H Feldman
- Division of Neurology, University of British Columbia, Vancouver, Canada; Department of Neurosciences, University of California, San Diego, La Jolla, USA
| | - Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Berteotti C, Liguori C, Pace M. Dysregulation of the orexin/hypocretin system is not limited to narcolepsy but has far-reaching implications for neurological disorders. Eur J Neurosci 2020; 53:1136-1154. [PMID: 33290595 DOI: 10.1111/ejn.15077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Neuropeptides orexin A and B (OX-A/B, also called hypocretin 1 and 2) are released selectively by a population of neurons which projects widely into the entire central nervous system but is localized in a restricted area of the tuberal region of the hypothalamus, caudal to the paraventricular nucleus. The OX system prominently targets brain structures involved in the regulation of wake-sleep state switching, and also orchestrates multiple physiological functions. The degeneration and dysregulation of the OX system promotes narcoleptic phenotypes both in humans and animals. Hence, this review begins with the already proven involvement of OX in narcolepsy, but it mainly discusses the new pre-clinical and clinical insights of the role of OX in three major neurological disorders characterized by sleep impairment which have been recently associated with OX dysfunction, such as Alzheimer's disease, stroke and Prader Willi syndrome, and have been emerged over the past 10 years to be strongly associated with the OX dysfunction and should be more considered in the future. In the light of the impairment of the OX system in these neurological disorders, it is conceivable to speculate that the integrity of the OX system is necessary for a healthy functioning body.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behaviour Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
19
|
Abstract
Twenty-two years after their discovery, the hypocretins (Hcrts), also known as orexins, are two of the most studied peptidergic systems, involved in myriad physiological systems that range from sleep, arousal, motivation, homeostatic regulation, fear, anxiety and learning. A causal relationship between activity of Hcrt and arousal stability was established shortly after their discovery and have led to the development of a new class of drugs to treat insomnia. In this review we discuss the many faces of the Hcrt system and examine recent findings that implicate decreased Hcrt function in the pathogenesis of a number of neuropsychiatric conditions. We also discuss future therapeutic strategies to replace or enhance Hcrt function as a treatment option for these neuropsychiatric conditions.
Collapse
Affiliation(s)
- Erica Seigneur
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
20
|
Um YH, Lim HK. Orexin and Alzheimer's Disease: A New Perspective. Psychiatry Investig 2020; 17:621-626. [PMID: 32517419 PMCID: PMC7385219 DOI: 10.30773/pi.2020.0136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Orexin's role in human cognition has recently been emphasized and emerging evidences indicate its close relationship with Alzheimer's disease (AD). This review aimed to demonstrate recent research on the relationship between orexin and AD. Orexin's role in stress regulation and memory is discussed, with significant findings related to sexual disparities in stress response, with potential clinical implications pertaining to AD pathology. There are controversies regarding the orexin levels in AD patients, but the role of orexin in the trajectory of AD is still emphasized in recent literatures. Orexin is also accentuated in the context of tau pathology, and orexin as a potential therapeutic target for AD is frequently discussed. Future directions with regard to the relationship between orexin and AD are suggested: 1) consideration for AD trajectory in the measurement of orexin levels, 2) the need for objective measure such as polysomnography and actigraphy, 3) the need for close observation of cognitive profiles of orexin-deficient narcolepsy patients, 4) the need for validation studies by neuroimaging 5) the need for taking account sexual disparities in orexinergic activiation, and 6) consideration for orexin's role as a stress regulator. The aforementioned new perspectives could help unravel the relationship between orexin and AD.
Collapse
Affiliation(s)
- Yoo Hyun Um
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
21
|
Grafe LA, Bhatnagar S. The contribution of orexins to sex differences in the stress response. Brain Res 2020; 1731:145893. [PMID: 30081036 PMCID: PMC6360123 DOI: 10.1016/j.brainres.2018.07.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/22/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023]
Abstract
Women are twice as likely as men to suffer from stress-related psychiatric disorders, such as post-traumatic stress disorder (PTSD) and Major Depressive Disorder (MDD), however, the biological basis of these sex differences is not fully understood. Interestingly, orexins are known to be dysregulated in these disorders. This review first discusses the important role of orexins regulating the response to stress. Next, we review the evidence for sex differences in the orexin system, in which the majority of both preclinical and clinical studies have reported higher orexin system expression in females. Finally, we discuss the functional consequences of these sex differences in orexin expression. Most importantly, the preclinical literature reveals that higher orexin system activity in females contributes to exaggerated neuroendocrine and behavioral responses to stress. In sum, the available data suggests that orexins may be important in the etiology of stress-related psychiatric disorders that present differently in men and women. Thus, targeting orexins could potentially ameliorate many phenotypes of stress-related illness in a sex-specific way.
Collapse
Affiliation(s)
- Laura A Grafe
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Vawter MP, Schulmann A, Alhassen L, Alhassen W, Hamzeh AR, Sakr J, Pauluk L, Yoshimura R, Wang X, Dai Q, Sanathara N, Civelli O, Alachkar A. Melanin Concentrating Hormone Signaling Deficits in Schizophrenia: Association With Memory and Social Impairments and Abnormal Sensorimotor Gating. Int J Neuropsychopharmacol 2020; 23:53-65. [PMID: 31563948 PMCID: PMC7442395 DOI: 10.1093/ijnp/pyz051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/30/2019] [Accepted: 09/24/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Evidence from anatomical, pharmacological, and genetic studies supports a role for the neuropeptide melanin concentrating hormone system in modulating emotional and cognitive functions. Genome-wide association studies revealed a potential association between the melanin concentrating hormone receptor (MCHR1) gene locus and schizophrenia, and the largest genome-wide association study conducted to date shows a credible genome-wide association. METHODS We analyzed MCHR1 and pro-melanin concentrating hormone RNA-Seq expression in the prefrontal cortex in schizophrenia patients and healthy controls. Disruptions in the melanin concentrating hormone system were modeled in the mouse brain by germline deletion of MCHR1 and by conditional ablation of melanin concentrating hormone expressing neurons using a Cre-inducible diphtheria toxin system. RESULTS MCHR1 expression is decreased in the prefrontal cortex of schizophrenia samples (false discovery rate (FDR) P < .05, CommonMind and PsychEncode combined datasets, n = 901) while pro-melanin concentrating hormone is below the detection threshold. MCHR1 expression decreased with aging (P = 6.6E-57) in human dorsolateral prefrontal cortex. The deletion of MCHR1 was found to lead to behavioral abnormalities mimicking schizophrenia-like phenotypes: hyperactivity, increased stereotypic and repetitive behavior, social impairment, impaired sensorimotor gating, and disrupted cognitive functions. Conditional ablation of pro-melanin concentrating hormone neurons increased repetitive behavior and produced a deficit in sensorimotor gating. CONCLUSIONS Our study indicates that early disruption of the melanin concentrating hormone system interferes with neurodevelopmental processes, which may contribute to the pathogenesis of schizophrenia. Further neurobiological research on the developmental timing and circuits that are affected by melanin concentrating hormone may lead to a therapeutic target for early prevention of schizophrenia.
Collapse
Affiliation(s)
- Marquis P Vawter
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, CA
| | - Anton Schulmann
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, CA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA
| | - Lamees Alhassen
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, CA
| | - Wedad Alhassen
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA
| | - Abdul Rezzak Hamzeh
- John Curtin School of Medical Research, Australian National University, Canberra
| | - Jasmine Sakr
- Department of Pharmaceutical Sciences, University of California, Irvine, CA
| | - Lucas Pauluk
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
| | - Ryan Yoshimura
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
| | - Xuejie Wang
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
| | - Qi Dai
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
| | - Nayna Sanathara
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
| | - Olivier Civelli
- Department of Pharmacology, School of Medicine, University of California, Irvine, CA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA
- Department of Developmental and Cell Biology, School of Medicine, University of California, Irvine, CA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, University of California, Irvine, CA
| |
Collapse
|
23
|
Abstract
Alzheimer's disease is a chronic neurodegenerative devastating disorder affecting a high percentage of the population over 65 years of age and causing a relevant emotional, social, and economic burden. Clinically, it is characterized by a prominent cognitive deficit associated with language and behavioral impairments. The molecular pathogenesis of Alzheimer's disease is multifaceted and involves changes in neurotransmitter levels together with alterations of inflammatory, oxidative, hormonal, and synaptic pathways, which may represent a drug target for both prevention and treatment; however, an effective treatment for Alzheimer's disease still represents an unmet goal. As neurotrophic factors participate in the modulation of the above-mentioned pathways, they have been highlighted as critical contributors of Alzheimer's disease etiology, whose modulation might be beneficial for Alzheimer's disease. We focused on the neurotrophin brain-derived neurotrophic factor, providing several lines of evidence pointing to brain-derived neurotrophic factor as a plausible endophenotype of cognitive deficits in Alzheimer's disease, illustrating some of the most recent possibilities to modulate the expression of this neurotrophin in the brain in an attempt to ameliorate cognition and delay the progression of Alzheimer's disease. This review shows that otherwise disparate pharmacologic or non-pharmacologic approaches converge on brain-derived neurotrophic factor, providing a means whereby apparently unrelated medical approaches may nevertheless produce similar synaptic and cognitive outcomes in Alzheimer's disease pathogenesis, suggesting that brain-derived neurotrophic factor-based synaptic repair may represent a modifying strategy to ameliorate cognition in Alzheimer's disease.
Collapse
|
24
|
Oh J, Eser RA, Ehrenberg AJ, Morales D, Petersen C, Kudlacek J, Dunlop SR, Theofilas P, Resende EDPF, Cosme C, Alho EJL, Spina S, Walsh CM, Miller BL, Seeley WW, Bittencourt JC, Neylan TC, Heinsen H, Grinberg LT. Profound degeneration of wake-promoting neurons in Alzheimer's disease. Alzheimers Dement 2019; 15:1253-1263. [PMID: 31416793 DOI: 10.1016/j.jalz.2019.06.3916] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/10/2019] [Accepted: 06/10/2019] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Sleep-wake disturbances are a common and early feature in Alzheimer's disease (AD). The impact of early tau pathology in wake-promoting neurons (WPNs) remains unclear. METHODS We performed stereology in postmortem brains from AD individuals and healthy controls to identify quantitative differences in morphological metrics in WPNs. Progressive supranuclear palsy (PSP) and corticobasal degeneration were included as disease-specific controls. RESULTS The three nuclei studied accumulate considerable amounts of tau inclusions and showed a decrease in neurotransmitter-synthetizing neurons in AD, PSP, and corticobasal degeneration. However, substantial neuronal loss was exclusively found in AD. DISCUSSION WPNs are extremely vulnerable to AD but not to 4 repeat tauopathies. Considering that WPNs are involved early in AD, such degeneration should be included in the models explaining sleep-wake disturbances in AD and considered when designing a clinical intervention. Sparing of WPNs in PSP, a condition featuring hyperinsomnia, suggest that interventions to suppress the arousal system may benefit patients with PSP.
Collapse
Affiliation(s)
- Jun Oh
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Rana A Eser
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Dulce Morales
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Cathrine Petersen
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jessica Kudlacek
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sara R Dunlop
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Panos Theofilas
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Elisa D P F Resende
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Celica Cosme
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Eduardo J L Alho
- Department of Neurology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Salvatore Spina
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Christine M Walsh
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jackson C Bittencourt
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Center for Neuroscience and Behavior, Institute of Psychology, University of Sao Paulo, Sao Paulo, Brazil
| | - Thomas C Neylan
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Helmut Heinsen
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Psychiatry, University of Wuerzburg, Wuerzburg, Germany
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
25
|
Oh ST, Liu QF, Jeong HJ, Lee S, Samidurai M, Jo J, Pak SC, Park HJ, Kim J, Jeon S. Nasal Cavity Administration of Melanin-Concentrating Hormone Improves Memory Impairment in Memory-Impaired and Alzheimer's Disease Mouse Models. Mol Neurobiol 2019; 56:8076-8086. [PMID: 31183806 DOI: 10.1007/s12035-019-01662-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
Melanin-concentrating hormone (MCH) is a highly conserved neuropeptide known to exhibit important functions in the brain. Some studies have reported that MCH improves memory by promoting memory retention. However, the precise molecular mechanisms by which MCH enhances memory impairment have yet to be fully elucidated. In this study, MCH was administered to the scopolamine-induced memory-impaired mice via the nasal cavity to examine the acute effects of MCH and Alzheimer's disease (AD) mouse models to evaluate the chronic effects of MCH. MCH improved memory impairment in both models and reduced soluble amyloid beta in the cerebral cortex of APP/PS1 transgenic mice. In vitro assays also showed that MCH inhibits amyloid beta-induced cytotoxicity. Furthermore, MCH increased long-term potentiation (LTP) in the hippocampus of wild-type and 5XFAD AD mouse model. To further elucidate the mechanisms of the chronic effect of MCH, the levels of phosphorylated CREB and GSK3β, and the expression of BDNF, TrkB and PSD95 were examined in the cerebral cortex and hippocampus. Our findings indicate that MCH might have neuroprotective effects via downstream pathways associated with the enhancement of neuronal synapses and LTP. This suggests a therapeutic potential of MCH for the treatment of neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Seung Tack Oh
- Research Institute, Dongkwang Pharmaceutical Company, Ltd., Seoul, 04535, Republic of Korea
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeongju, 38066, Republic of Korea
| | - Ha Jin Jeong
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, 04933, Republic of Korea
| | - Seongmi Lee
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, 04933, Republic of Korea
| | - Manikandan Samidurai
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jihoon Jo
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.,NeuroMedical Convergence Lab, Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, New South Wales, 2795, Australia
| | - Hi-Joon Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea. .,Integrative Parkinson's Disease Research Group, Studies of Translational Acupuncture Research (STAR), Acupuncture & Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Songhee Jeon
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
26
|
Cengiz M, Karaj V, Kocabasoğlu N, Gozubatik-Celik G, Dirican A, Bayoglu B. Orexin/hypocretin receptor, Orx1, gene variants are associated with major depressive disorder. Int J Psychiatry Clin Pract 2019; 23:114-121. [PMID: 30596528 DOI: 10.1080/13651501.2018.1551549] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: Orexins (hypocretins) are neuropeptides expressed in hypothalamic neurons and have regulatory roles in feeding/drinking behaviours, endocrine functions and sleep/wakefulness state. Major depressive disorder (MDD) is a major mood disorder and neurotransmitter dysfunction in hypothalamic neurons may have roles in its formation. Hence, we conducted experiments to determine whether orexin receptor 1 and 2 (Orx1, Orx2) genes were associated with MDD development. Methods: Seventy-five MDD patients and 87 healthy controls were enrolled for the study. Genotyping was carried out with real-time polymerase chain reaction (RT-PCR). Hamilton Rating-Scale for Depression (HRSD) and Beck Depression Inventory (BDI) were utilized to evaluate depressive symptom severity. Results: A significant relation was found in genotype frequencies of Orx1 rs10914456 and rs2271933 variants between MDD patients and controls (p = .009, p = .006). Rs10914456 CC genotype increased MDD risk 3.57 times more than carrying other genotypes (p = .008, OR =3.57;95% CI: 1.39-9.14). However, no association was observed in Orx2 rs2653349 genotypes for MDD development (p > .05). Although statistically not significant, HRSD scores were diminished in MDD subjects carrying rs10914456 CC variants when compared with CT and TT variants (p = .069). Conclusion. This study suggests that, Orx1 rs10914456 and rs2271933 can be associated with MDD development. Hence, Orx1 rs10914456 variants may affect depressive symptom severity.
Collapse
Affiliation(s)
- Mujgan Cengiz
- a Department of Medical Biology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Vilson Karaj
- b Department of Science, Institute of Forensic Sciences , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Nese Kocabasoğlu
- c Department of Psychiatry, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Gokcen Gozubatik-Celik
- d Department of Neurology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| | - Ahmet Dirican
- e Department of Biostatistics and Medical Informatics, Istanbul Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Burcu Bayoglu
- a Department of Medical Biology, Cerrahpasa Medical Faculty , Istanbul University-Cerrahpasa , Istanbul , Turkey
| |
Collapse
|
27
|
Gabelle A, Jaussent I, Bouallègue FB, Lehmann S, Lopez R, Barateau L, Grasselli C, Pesenti C, de Verbizier D, Béziat S, Mariano-Goulart D, Carlander B, Dauvilliers Y. Reduced brain amyloid burden in elderly patients with narcolepsy type 1. Ann Neurol 2018; 85:74-83. [PMID: 30387527 DOI: 10.1002/ana.25373] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To determine whether brain amyloid burden in elderly patients with narcolepsy type 1 (NT1) is lower than in controls, and to assess in patients with NT1 the relationships between amyloid burden, cerebral spinal fluid (CSF) markers of Alzheimer disease (AD), CSF orexin-A, and cognitive profile. METHODS Cognitive and 18 F-florbetapir positron emission tomography (PET) data were compared in patients with NT1 aged ≥ 65 years (n = 23) and in age- and sex-matched controls free of clinical dementia selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI; n = 69) and the Multi-Domain Intervention Alzheimer's Prevention Trial (MAPT-18F AV45-PET; n = 23) cohorts. The standardized uptake values (SUVs) of the cortical retention index for 6 regions of interest were computed and averaged to create a mean SUV ratio normalized to 3 subcortical reference regions (cerebellum, pons, and a composite region). A cortical/cerebellum SUV ratio ≥ 1.17 defined positive PET amyloid. RESULTS Lower cortical amyloid burden was observed in the NT1 than in the ADNI and MAPT-AV45 groups (mean cortical/cerebellum SUV ratios = 0.95 ± 0.15, 1.11 ± 0.18 [p < 0.0001], and 1.14 ± 0.17 [p = 0.0005], respectively). Similar results were obtained with all subcortical reference regions and for all cortical regions of interest, except cingulum. Only 1 patient with NT1 (4.4%) had positive PET amyloid compared with 27.5% in the ADNI and 30.4% in the MAPT-AV45 group. In the NT1 group, cortical or regional amyloid load was not associated with CSF orexin-A, CSF AD biomarkers, or neuropsychological profile. INTERPRETATION Lower brain amyloid burden, assessed by 18 F-florbetapir PET, in patients with NT1 suggests delayed appearance of amyloid plaques. ANN NEUROL 2019;85:74-83.
Collapse
Affiliation(s)
- Audrey Gabelle
- Memory Research and Resources Center, Department of Neurology, Gui de Chauliac University Hospital Center.,University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research
| | - Isabelle Jaussent
- University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research
| | - Fayçal Ben Bouallègue
- University of Montpellier.,Department of Nuclear Medicine, Montpellier University Hospital.,PhyMedExp, National Institute of Health and Medical Research, National Center for Scientific Research
| | - Sylvain Lehmann
- University of Montpellier.,National Institute of Health and Medical Research U1183, Saint Eloi Hospital
| | - Régis Lopez
- University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research.,Narcolepsy National Reference Center, Sleep Center, Department of Neurology, Montpellier University Hospital Center, Montpellier, France
| | - Lucie Barateau
- University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research.,Narcolepsy National Reference Center, Sleep Center, Department of Neurology, Montpellier University Hospital Center, Montpellier, France
| | - Caroline Grasselli
- Memory Research and Resources Center, Department of Neurology, Gui de Chauliac University Hospital Center
| | - Carole Pesenti
- Narcolepsy National Reference Center, Sleep Center, Department of Neurology, Montpellier University Hospital Center, Montpellier, France
| | | | - Séverine Béziat
- University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research
| | - Denis Mariano-Goulart
- University of Montpellier.,Department of Nuclear Medicine, Montpellier University Hospital.,PhyMedExp, National Institute of Health and Medical Research, National Center for Scientific Research
| | - Bertrand Carlander
- Narcolepsy National Reference Center, Sleep Center, Department of Neurology, Montpellier University Hospital Center, Montpellier, France
| | - Yves Dauvilliers
- University of Montpellier.,National Institute of Health and Medical Research U1061, Neuropsychiatry: Epidemiological and Clinical Research.,Narcolepsy National Reference Center, Sleep Center, Department of Neurology, Montpellier University Hospital Center, Montpellier, France
| | | | | |
Collapse
|
28
|
Hiller AJ, Ishii M. Disorders of Body Weight, Sleep and Circadian Rhythm as Manifestations of Hypothalamic Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2018; 12:471. [PMID: 30568576 PMCID: PMC6289975 DOI: 10.3389/fncel.2018.00471] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022] Open
Abstract
While cognitive decline and memory loss are the major clinical manifestations of Alzheimer’s disease (AD), they are now recognized as late features of the disease. Recent failures in clinical drug trials highlight the importance of evaluating and treating patients with AD as early as possible and the difficulties in developing effective therapies once the disease progresses. Since the pathological hallmarks of AD including the abnormal aggregation of amyloid-beta (Aβ) and tau can occur decades before any significant cognitive decline in the preclinical stage of AD, it is important to identify the earliest clinical manifestations of AD and elucidate their underlying cellular and molecular mechanisms. Importantly, metabolic and non-cognitive manifestations of AD such as weight loss and alterations of peripheral metabolic signals can occur before the onset of cognitive symptoms and worsen with disease progression. Accumulating evidence suggests that the major culprit behind these early metabolic and non-cognitive manifestations of AD is AD pathology causing dysfunction of the hypothalamus, a brain region critical for integrating peripheral signals with essential homeostatic physiological functions. Here, we aim to highlight recent developments that address the role of AD pathology in the development of hypothalamic dysfunction associated with metabolic and non-cognitive manifestations seen in AD. Understanding the mechanisms underlying hypothalamic dysfunction in AD could give key new insights into the development of novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Abigail J Hiller
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Makoto Ishii
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States.,Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
29
|
Abstract
The neuropeptides orexins are important in regulating the neurobiological systems that respond to stressful stimuli. Furthermore, orexins are known to play a role many of the phenotypes associated with stress-related mental illness such as changes in cognition, sleep-wake states, and appetite. Interestingly, orexins are altered in stress-related psychiatric disorders such as Major Depressive Disorder and Anxiety Disorders. Thus, orexins may be a potential target for treatment of these disorders. In this review, we will focus on what is known about the role of orexins in acute and repeated stress, in stress-induced phenotypes relevant to psychiatric illness in preclinical models, and in stress-related psychiatric illness in humans. We will also briefly discuss how orexins may contribute to sex differences in the stress response and subsequent phenotypes relevant to mental health, as many stress-related psychiatric disorders are twice as prevalent in women.
Collapse
|
30
|
|
31
|
Sil’kis IG. A Neurochemical Approach to the Search for Drugs for the Treatment of Symptoms of Alzheimer’s Disease. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Peineau S, Rabiant K, Pierrefiche O, Potier B. Synaptic plasticity modulation by circulating peptides and metaplasticity: Involvement in Alzheimer's disease. Pharmacol Res 2018; 130:385-401. [PMID: 29425728 DOI: 10.1016/j.phrs.2018.01.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 10/18/2022]
Abstract
Synaptic plasticity is a cellular process involved in learning and memory whose alteration in its two main forms (Long Term Depression (LTD) and Long Term Potentiation (LTP)), is observed in most brain pathologies, including neurodegenerative disorders such as Alzheimer's disease (AD). In humans, AD is associated at the cellular level with neuropathological lesions composed of extracellular deposits of β-amyloid (Aβ) protein aggregates and intracellular neurofibrillary tangles, cellular loss, neuroinflammation and a general brain homeostasis dysregulation. Thus, a dramatic synaptic environment perturbation is observed in AD patients, involving changes in brain neuropeptides, cytokines, growth factors or chemokines concentration and diffusion. Studies performed in animal models demonstrate that these circulating peptides strongly affect synaptic functions and in particular synaptic plasticity. Besides this neuromodulatory action of circulating peptides, other synaptic plasticity regulation mechanisms such as metaplasticity are altered in AD animal models. Here, we will review new insights into the study of synaptic plasticity regulatory/modulatory mechanisms which could influence the process of synaptic plasticity in the context of AD with a particular attention to the role of metaplasticity and peptide dependent neuromodulation.
Collapse
Affiliation(s)
- Stéphane Peineau
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France; Centre for Synaptic Plasticity, School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.
| | - Kevin Rabiant
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France
| | - Olivier Pierrefiche
- GRAP UMR1247, INSERM, Centre Universitaire de Recherche en Santé, Université de Picardie Jules Verne, Amiens, France.
| | - Brigitte Potier
- Laboratoire Aimé Cotton, CNRS-ENS UMR9188, Université Paris-Sud, Orsay, France.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Spontaneous physical activity (SPA) is a physical activity not motivated by a rewarding goal, such as that associated with food-seeking or wheel-running behavior. SPA is often thought of as only "fidgeting," but that is a mischaracterization, since fidgety behavior can be linked to stereotypies in neurodegenerative disease and other movement disorders. Instead, SPA should be thought of as all physical activity behavior that emanates from an unconscious drive for movement. RECENT FINDINGS An example of this may be restless behavior, which can include fidgeting and gesticulating, frequent sit-to-stand movement, and more time spent standing and moving. All physical activity burns calories, and as such, SPA could be manipulated as a means to burn calories, and defend against weight gain and reduce excess adiposity. In this review, we discuss human and animal literature on the use of SPA in reducing weight gain, the neuromodulators that could be targeted to this end, and future directions in this field.
Collapse
Affiliation(s)
- Catherine M Kotz
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA.
- GRECC, Minneapolis VA Health Care System, GRECC, One Veterans Drive, Minneapolis, MN, 55417, USA.
| | | | - Jennifer A Teske
- Department of Nutritional Sciences, University of Arizona, 1177 E 4th street, Shantz 332, Tucson, AZ, 85721, USA
| | - Charles J Billington
- Department of Medicine, University of Minnesota, 420 Delaware St SE, Minneapolis, MN, 5545, USA
- Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN, 55417, USA
| |
Collapse
|
34
|
Bubu OM, Brannick M, Mortimer J, Umasabor-Bubu O, Sebastião YV, Wen Y, Schwartz S, Borenstein AR, Wu Y, Morgan D, Anderson WM. Sleep, Cognitive impairment, and Alzheimer's disease: A Systematic Review and Meta-Analysis. Sleep 2017; 40:2661823. [PMID: 28364458 DOI: 10.1093/sleep/zsw032] [Citation(s) in RCA: 366] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2016] [Indexed: 12/24/2022] Open
Abstract
Study Objectives Mounting evidence implicates disturbed sleep or lack of sleep as one of the risk factors for Alzheimer's disease (AD), but the extent of the risk is uncertain. We conducted a broad systematic review and meta-analysis to quantify the effect of sleep problems/disorders on cognitive impairment and AD. Methods Original published literature assessing any association of sleep problems or disorders with cognitive impairment or AD was identified by searching PubMed, Embase, Web of Science, and the Cochrane library. Effect estimates of individual studies were pooled and relative risks (RR) and 95% confidence intervals (CI) were calculated using random effects models. We also estimated the population attributable risk. Results Twenty-seven observational studies (n = 69216 participants) that provided 52 RR estimates were included in the meta-analysis. Individuals with sleep problems had a 1.55 (95% CI: 1.25-1.93), 1.65 (95% CI: 1.45-1.86), and 3.78 (95% CI: 2.27-6.30) times higher risk of AD, cognitive impairment, and preclinical AD than individuals without sleep problems, respectively. The overall meta-analysis revealed that individuals with sleep problems had a 1.68 (95% CI: 1.51-1.87) times higher risk for the combined outcome of cognitive impairment and/or AD. Approximately 15% of AD in the population may be attributed to sleep problems. Conclusion This meta-analysis confirmed the association between sleep and cognitive impairment or AD and, for the first time, consolidated the evidence to provide an "average" magnitude of effect. As sleep problems are of a growing concern in the population, these findings are of interest for potential prevention of AD.
Collapse
Affiliation(s)
- Omonigho M Bubu
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Michael Brannick
- Psychology Department, College of Arts and Sciences, University of South Florida, Tampa, FL
| | - James Mortimer
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Ogie Umasabor-Bubu
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Yuri V Sebastião
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Yi Wen
- Department of Chemical and Biomedical Engineering, College of Engineering, University of South Florida, Tampa, FL
| | - Skai Schwartz
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Amy R Borenstein
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Yougui Wu
- Department of Epidemiology & Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - David Morgan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL.,Byrd Alzheimer Institute, Tampa, FL
| | - William M Anderson
- Sleep Medicine and Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| |
Collapse
|
35
|
Abstract
Alzheimer's disease (AD) is the most frequent age-related dementia. It prevalently causes cognitive decline, although it is frequently associated with secondary behavioral disturbances. AD neurodegeneration characteristically produces a remarkable destruction of the sleep-wake cycle, with diurnal napping, nighttime arousals, sleep fragmentation, and REM sleep impairment. It was recently hypothesized that the orexinergic system was involved in AD pathology. Accordingly, recent papers showed the association between orexinergic neurotransmission dysfunction, sleep impairment, and cognitive decline in AD. Orexin is a hypothalamic neurotransmitter which physiologically produces wakefulness and reduces REM sleep and may alter the sleep-wake cycle in AD patients. Furthermore, the orexinergic system seems to interact with CSF AD biomarkers, such as beta-amyloid and tau proteins. Beta-amyloid accumulation is the main hallmark of AD pathology, while tau proteins mark brain neuronal injury due to AD pathology. Investigations so far suggest that orexinergic signaling overexpression alters the sleep-wake cycle and secondarily induces beta-amyloid accumulation and tau-mediated neurodegeneration. Therefore, considering that orexinergic system dysregulation impairs sleep-wake rhythms and may influence AD pathology, it is hypothesized that orexin receptor antagonists are likely potential preventive/therapeutic options in AD patients.
Collapse
Affiliation(s)
- Claudio Liguori
- Sleep Medicine Centre, Neurophysiopathology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
36
|
Raman NM, Ramasamy S. Genetic validation and spectroscopic detailing of DHN-melanin extracted from an environmental fungus. Biochem Biophys Rep 2017; 12:98-107. [PMID: 28955797 PMCID: PMC5613234 DOI: 10.1016/j.bbrep.2017.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 11/30/2022] Open
Abstract
Accurate characterization of melanin using analytical methodologies has proved to be difficult due to its heterogeneity, insolubility in wide pH and broad range of solvents. The present study was undertaken to characterize melanin extracted from an environmental Aspergillus fumigatus AFGRD105 by studying its genes, chemical properties and spectral data. A gene based approach to confirm the type of melanin carried out indicated the extracted melanin to be of the dihydroxynaphthalene type. On comparison with synthetic melanin, UV–Vis and IR spectra of the extracted melanin revealed characteristic peaks that can be further used for confirmation of DHN-melanin extracted from any source. Solid state 13C NMR spectroscopy established the presence of the hydroxyl-naphthalene moiety and validated the results obtained by genetic analysis. The correct assignment of the observed spectral frequency characteristic of functional groups can be further adapted in future works that deal with binding capacities and biomolecule systems involving melanin. DNA was extracted by a standardised protocol that can be adapted for environmental and clinically isolated fungi. The presence of genes was used to identify the type of melanin. Physico chemical characterization of the melanin extracted was performed. UV–Vis and IR spectra were used to confirm the type of melanin. Further, the chemical moieties were substantiated using 13C solid state NMR spectroscopy.
Collapse
Affiliation(s)
- Nitya Meenakshi Raman
- Department of Biotechnology, Dr. G. R. Damodaran College of Science, Bharathiar University, Coimbatore 641014, India
| | - Suganthi Ramasamy
- Department of Biotechnology, Dr. G. R. Damodaran College of Science, Bharathiar University, Coimbatore 641014, India
| |
Collapse
|
37
|
Al Suwayri SM, BaHammam AS. The "Known Unknowns" of Kleine-Levin Syndrome: A Review and Future Prospects. Sleep Med Clin 2017; 12:345-358. [PMID: 28778233 DOI: 10.1016/j.jsmc.2017.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Kleine-Levin syndrome (KLS) is a rare, homogeneous, debilitating sleep disorder characterized by episodic hypersomnia, cognitive impairment, and behavioral changes. The etiology, pathophysiology, and optimal management of KLS remain uncertain. We identify the 5 key areas requiring urgent attention: KLS immunopathogenesis studies, next-generation genetics, multimodal functional imaging, biomarker discovery, and clinical drug trials. A centralized registry of afflicted individuals must be established. Disease uniformity should make the identification of associated genetic or imaging biomarkers easier, but clinical efforts require laboratory-based research to model the disease and generate preclinical data for clinical translation.
Collapse
Affiliation(s)
- Saad M Al Suwayri
- Department of Internal Medicine, College of Medicine, Al Imam Mohammad Ibn Saud Islamic University (IMSIU), PO Box 7544, Othman bin Afan Road, Riyadh, Saudi Arabia
| | - Ahmed S BaHammam
- University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Box 225503, Riyadh 11324, Saudi Arabia.
| |
Collapse
|
38
|
Silkis IG. Hypothetical neurochemical mechanisms of paradoxical sleep deficiency in Alzheimer’s disease. NEUROCHEM J+ 2017. [DOI: 10.1134/s181971241702012x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
39
|
Kent BA, Mistlberger RE. Sleep and hippocampal neurogenesis: Implications for Alzheimer's disease. Front Neuroendocrinol 2017; 45:35-52. [PMID: 28249715 DOI: 10.1016/j.yfrne.2017.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/23/2017] [Accepted: 02/24/2017] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and currently there are no effective disease-modifying treatments available. Hallmark symptoms of AD include impaired hippocampus-dependent episodic memory and disrupted sleep and circadian rhythms. The pathways connecting these symptoms are of particular interest because it is well established that sleep and circadian disruption can impair hippocampus-dependent learning and memory. In rodents, these procedures also markedly suppress adult hippocampal neurogenesis, a form of brain plasticity that is believed to play an important role in pattern separation, and thus episodic memory. A causal role for sleep disruptions in AD pathophysiology is suggested by evidence for sleep-dependent glymphatic clearance of metabolic waste products from the brain. This review explores a complementary hypothesis that sleep and circadian disruptions in AD contribute to cognitive decline by activating neuroendocrine and neuroinflammatory signaling pathways that suppress hippocampal neurogenesis. Evidence for this hypothesis underscores the promise of sleep, circadian rhythms, and neurogenesis as therapeutic targets for remediation of memory impairment in AD.
Collapse
Affiliation(s)
- Brianne A Kent
- Division of Neurology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
40
|
Cedernaes J, Osorio RS, Varga AW, Kam K, Schiöth HB, Benedict C. Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer's disease. Sleep Med Rev 2017; 31:102-111. [PMID: 26996255 PMCID: PMC4981560 DOI: 10.1016/j.smrv.2016.02.002] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 12/13/2022]
Abstract
During wakefulness, extracellular levels of metabolites in the brain increase. These include amyloid beta (Aβ), which contributes to the pathogenesis of Alzheimer's disease (AD). Counterbalancing their accumulation in the brain, sleep facilitates the removal of these metabolites from the extracellular space by convective flow of the interstitial fluid from the para-arterial to the para-venous space. However, when the sleep-wake cycle is disrupted (characterized by increased brain levels of the wake-promoting neuropeptide orexin and increased neural activity), the central nervous system (CNS) clearance of extracellular metabolites is diminished. Disruptions to the sleep-wake cycle have furthermore been linked to increased neuronal oxidative stress and impaired blood-brain barrier function - conditions that have also been proposed to play a role in the development and progression of AD. Notably, recent human and transgenic animal studies have demonstrated that AD-related pathophysiological processes that occur long before the clinical onset of AD, such as Aβ deposition in the brain, disrupt sleep and circadian rhythms. Collectively, as proposed in this review, these findings suggest the existence of a mechanistic interplay between AD pathogenesis and disrupted sleep-wake cycles, which is able to accelerate the development and progression of this disease.
Collapse
Affiliation(s)
| | - Ricardo S Osorio
- Center for Brain Health, NYU Langone Medical Center, New York, NY, USA.
| | - Andrew W Varga
- NYU Sleep Disorders Center, NYU Langone Medical Center, New York, NY, USA
| | - Korey Kam
- NYU Sleep Disorders Center, NYU Langone Medical Center, New York, NY, USA
| | | | | |
Collapse
|
41
|
Cerebrospinal fluid levels of orexin-A and histamine, and sleep profile within the Alzheimer process. Neurobiol Aging 2017; 53:59-66. [PMID: 28235679 DOI: 10.1016/j.neurobiolaging.2017.01.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 01/07/2017] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
To better understand how sleep/wake dysregulation affects Alzheimer's disease (AD), we compared the cerebrospinal fluid (CSF) orexin and histamine/tele-methylhistamine (HA/t-MHA) levels of 82 patients (41 probable-AD-high level of evidence, 41 mild cognitive impairment MCI-due-to-AD), 24 other neurologic disorders (OND) and 24 controls. We determined the relationships between these biomarkers, the CSF AD biomarkers concentrations, and the clinical sleep profile. CSF orexin-A but not HA/t-MHA levels were higher in MCI and AD than OND and controls. CSF orexin-A is correlated to CSF amyloid-β42in MCI and AD, independently of age, gender, MMSE, total-tau/phosphorylated-tau, HA or sleep parameters. Nighttime sleep duration was longer in MCI and AD patients than controls. In MCI, nighttime sleep duration negatively correlated with CSF amyloid-β42 and MMSE. To conclude, CSF orexin-A but not HA/t-HMA was upregulated in AD and correlated with amyloid-β42 level. Our data suggested a change in the sleep-wake pattern at an early stage of the disease that needs further investigation to deeply explain the mechanistic interplay between sleep and Alzheimer.
Collapse
|
42
|
The choroid plexus as a sex hormone target: Functional implications. Front Neuroendocrinol 2017; 44:103-121. [PMID: 27998697 DOI: 10.1016/j.yfrne.2016.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/25/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022]
Abstract
The choroid plexuses (CPs) are highly vascularized branched structures that protrude into the ventricles of the brain, and form a unique interface between the blood and the cerebrospinal fluid (CSF). In recent years, novel functions have been attributed to this tissue such as in immune and chemical surveillance of the central nervous system, brain development, adult neurogenesis and circadian rhythm regulation. Sex hormones (SH) are widely recognized as modulators in several neurodegenerative diseases, and there is evidence that estrogens and androgens regulate several fundamental biological functions in the CPs. Therefore, SH are likely to affect the composition of the CSF impacting on brain homeostasis. This review will look at implications of the CPs' sex-related specificities.
Collapse
|
43
|
Villa C, Ferini-Strambi L, Combi R. The Synergistic Relationship between Alzheimer's Disease and Sleep Disorders: An Update. J Alzheimers Dis 2016; 46:571-80. [PMID: 25835421 DOI: 10.3233/jad-150138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Sleep disorders are frequently reported in Alzheimer's disease (AD), with a significant impact on patients and caregivers and a major risk factor for early institutionalization. Although changes in sleep organization are a hallmark of the normal aging processes, sleep macro- and micro-architectural alterations are more evident in patients affected by AD. Degeneration of neural pathways regulating sleep-wake patterns and sleep architecture may contribute to sleep alterations. In return, several recent studies suggested that common sleep disorders may precede clinical symptoms of dementia and represent risk factors for cognitive decline, through impairment of sleep-dependent memory consolidation processes. Thus, a close relationship between sleep disorders and AD has been largely hypothesized. Here, sleep alterations in AD and its pre-dementia stage, mild cognitive impairment, and their complex interactions are reviewed.
Collapse
Affiliation(s)
- Chiara Villa
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy
| | - Luigi Ferini-Strambi
- Department of Clinical Neurosciences, Sleep Disorders Center, University Vita-Salute San Raffaele, Milan, Italy
| | - Romina Combi
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
44
|
Osorio RS, Ducca EL, Wohlleber ME, Tanzi EB, Gumb T, Twumasi A, Tweardy S, Lewis C, Fischer E, Koushyk V, Cuartero-Toledo M, Sheikh MO, Pirraglia E, Zetterberg H, Blennow K, Lu SE, Mosconi L, Glodzik L, Schuetz S, Varga AW, Ayappa I, Rapoport DM, de Leon MJ. Orexin-A is Associated with Increases in Cerebrospinal Fluid Phosphorylated-Tau in Cognitively Normal Elderly Subjects. Sleep 2016; 39:1253-60. [PMID: 26951396 DOI: 10.5665/sleep.5846] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/07/2016] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES To evaluate the role of orexin-A with respect to cerebrospinal fluid (CSF) Alzheimer disease (AD) biomarkers, and explore its relationship to cognition and sleep characteristics in a group of cognitively normal elderly individuals. METHODS Subjects were recruited from multiple community sources for National Institutes of Health supported studies on normal aging, sleep and CSF biomarkers. Sixty-three participants underwent home monitoring for sleep-disordered breathing, clinical, sleep and cognitive evaluations, as well as a lumbar puncture to obtain CSF. Individuals with medical history or with magnetic resonance imaging evidence of disorders that may affect brain structure or function were excluded. Correlation and linear regression analyses were used to assess the relationship between orexin-A and CSF AD-biomarkers controlling for potential sociodemographic and sleep confounders. RESULTS Levels of orexin-A, amyloid beta 42 (Aβ42), phosphorylated-tau (P-Tau), total-tau (T-Tau), Apolipoprotein E4 status, age, years of education, reported total sleep time, number of awakenings, apnea-hypopnea indices (AHI), excessive daytime sleepiness, and a cognitive battery were analyzed. Subjects were 69.59 ± 8.55 years of age, 57.1% were female, and 30.2% were apolipoprotein E4+. Orexin-A was positively correlated with Aβ42, P-Tau, and T-Tau. The associations between orexin-A and the AD-biomarkers were driven mainly by the relationship between orexin-A and P-Tau and were not influenced by other clinical or sleep characteristics that were available. CONCLUSIONS Orexin-A is associated with increased P-Tau in normal elderly individuals. Increases in orexin-A and P-Tau might be a consequence of the reduction in the proportion of the deeper, more restorative slow wave sleep and rapid eye movement sleep reported with aging. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov registration number NCT01962779.
Collapse
Affiliation(s)
| | - Emma L Ducca
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY
| | | | - Emily B Tanzi
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Tyler Gumb
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Akosua Twumasi
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Samuel Tweardy
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Clifton Lewis
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Esther Fischer
- Department of Neuroscience, JFK Medical Center, Edison, NJ
| | | | | | | | | | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Shou-En Lu
- Department of Biostatistics, Rutgers School of Public Health, Piscataway, NJ
| | - Lisa Mosconi
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Lidia Glodzik
- Center for Brain Health, NYU School of Medicine, New York, NY
| | - Sonja Schuetz
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY
| | - Andrew W Varga
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY
| | - Indu Ayappa
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY
| | - David M Rapoport
- Division of Pulmonary, Critical Care, and Sleep Medicine, NYU School of Medicine, New York, NY
| | - Mony J de Leon
- Center for Brain Health, NYU School of Medicine, New York, NY
| |
Collapse
|
45
|
Hirtz C, Vialaret J, Gabelle A, Nowak N, Dauvilliers Y, Lehmann S. From radioimmunoassay to mass spectrometry: a new method to quantify orexin-A (hypocretin-1) in cerebrospinal fluid. Sci Rep 2016; 6:25162. [PMID: 27165941 PMCID: PMC4863245 DOI: 10.1038/srep25162] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/11/2016] [Indexed: 12/14/2022] Open
Abstract
I125 radioimmunoassay (RIA) is currently the standard technique for quantifying cerebrospinal fluid (CSF) orexin-A/hypocretin-1, a biomarker used to diagnose narcolepsy type 1. However, orexin-A RIA is liable to undergo cross-reactions with matrix constituents generating interference, high variability between batches, low precision and accuracy, and requires special radioactivity precautions. Here we developed the first quantitative mass spectrometry assay of orexin-A based on a multiple reaction monitoring (MRM) approach. This method was tested in keeping with the Clinical and Laboratory Standards Institute (CLSI) guidelines and its clinical relevance was confirmed by comparing patients with narcolepsy type 1 versus patients with other neurological conditions. The results obtained using MRM and RIA methods were highly correlated, and Bland–Altman analysis established their interchangeability. However, the MRM values had a wider distribution and were 2.5 time lower than the RIA findings. In conclusion, this method of assay provides a useful alternative to RIA to quantify orexin-A, and may well replace it not only in narcolepsy type 1, but also in the increasing number of pathologies in which the quantification of this analyte is relevant.
Collapse
Affiliation(s)
- Christophe Hirtz
- CHU Montpellier, Institut de Recherche en Biothérapie, hôpital St Eloi, Laboratoire de Biochimie Protéomique Clinique et CRB, Montpellier, F-34000 France.,Université de Montpellier, Montpellier, F-34000 France. INSERM U1183, Montpellier, F-34000 France
| | - Jérôme Vialaret
- CHU Montpellier, Institut de Recherche en Biothérapie, hôpital St Eloi, Laboratoire de Biochimie Protéomique Clinique et CRB, Montpellier, F-34000 France.,Université de Montpellier, Montpellier, F-34000 France. INSERM U1183, Montpellier, F-34000 France
| | - Audrey Gabelle
- CHU Montpellier, Institut de Recherche en Biothérapie, hôpital St Eloi, Laboratoire de Biochimie Protéomique Clinique et CRB, Montpellier, F-34000 France.,Université de Montpellier, Montpellier, F-34000 France. INSERM U1183, Montpellier, F-34000 France.,Memory Research Resources center, Department of Neurology, Gui-de-Chauliac Hospital, Montpellier University Hospital, F-34000 France
| | - Nora Nowak
- CHU Montpellier, Institut de Recherche en Biothérapie, hôpital St Eloi, Laboratoire de Biochimie Protéomique Clinique et CRB, Montpellier, F-34000 France.,Université de Montpellier, Montpellier, F-34000 France. INSERM U1183, Montpellier, F-34000 France
| | - Yves Dauvilliers
- National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic hypersomnia and Kleine-Levin Syndrome, France.,Sleep Disorders Center, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, Inserm U1061, Montpellier, France
| | - Sylvain Lehmann
- CHU Montpellier, Institut de Recherche en Biothérapie, hôpital St Eloi, Laboratoire de Biochimie Protéomique Clinique et CRB, Montpellier, F-34000 France.,Université de Montpellier, Montpellier, F-34000 France. INSERM U1183, Montpellier, F-34000 France
| |
Collapse
|
46
|
Rapid eye movement sleep disruption and sleep fragmentation are associated with increased orexin-A cerebrospinal-fluid levels in mild cognitive impairment due to Alzheimer's disease. Neurobiol Aging 2016; 40:120-126. [DOI: 10.1016/j.neurobiolaging.2016.01.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 01/30/2023]
|
47
|
Urrestarazu E, Iriarte J. Clinical management of sleep disturbances in Alzheimer's disease: current and emerging strategies. Nat Sci Sleep 2016; 8:21-33. [PMID: 26834500 PMCID: PMC4716729 DOI: 10.2147/nss.s76706] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sleep and circadian disorders in Alzheimer's disease (AD) are more frequent than in the general population and appear early in the course of the disease. Quality of sleep and quality of life are parallel in these patients, and such disorders also represent a heavy burden for caregivers. Although alterations in melatonin and hypocretins (orexins) seem to play a key role in the origin of these disturbances, the etiology of these disorders is multifactorial, including many factors such as environment, behavior, treatments, and comorbidities, among others. A comprehensive evaluation of sleep in each patient is essential in the design of the treatment that includes nonpharmacological and pharmacological approaches. One particularly interesting point is the possibility of a role of sleep disorders in the pathogenesis of AD, raising the possibility that treating the sleep disorder may alter the course of the disease. In this review, we present an update on the role of sleep disorders in AD, the bidirectional influence of sleep problems and AD, and treatment options. Behavioral measures, bright light therapy (BLT), melatonin, and other drugs are likely well known and correctly managed by the physicians in charge of these patients. In spite of the multiple treatments used, evidence of efficacy is scarce and more randomized double-blind placebo-controlled studies are needed. Future directions for treatment are the establishment of BLT protocols and the development of drugs with new mechanisms of action, especially hypocretin receptor antagonists, melatonin receptor agonists, and molecules that modulate the circadian clock.
Collapse
Affiliation(s)
- Elena Urrestarazu
- Sleep Unit, Clinical Neurophysiology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Jorge Iriarte
- Sleep Unit, Clinical Neurophysiology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
48
|
Ishii M, Iadecola C. Metabolic and Non-Cognitive Manifestations of Alzheimer's Disease: The Hypothalamus as Both Culprit and Target of Pathology. Cell Metab 2015; 22:761-76. [PMID: 26365177 PMCID: PMC4654127 DOI: 10.1016/j.cmet.2015.08.016] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is increasingly recognized as a complex neurodegenerative disease beginning decades prior to the cognitive decline. While cognitive deficits remain the cardinal manifestation of AD, metabolic and non-cognitive abnormalities, such as alterations in body weight and neuroendocrine functions, are also present, often preceding the cognitive decline. Furthermore, hypothalamic dysfunction can also be a driver of AD pathology. Here we offer a brief appraisal of hypothalamic dysfunction in AD and provide insight into an underappreciated dual role of the hypothalamus as both a culprit and target of AD pathology, as well as into new opportunities for therapeutic interventions and biomarker development.
Collapse
Affiliation(s)
- Makoto Ishii
- Feil Family Brain and Mind Research Institute, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY 10065, USA
| |
Collapse
|
49
|
Johansson P, Almqvist EG, Wallin A, Johansson JO, Andreasson U, Blennow K, Zetterberg H, Svensson J. Cerebrospinal fluid substance P concentrations are elevated in patients with Alzheimer's disease. Neurosci Lett 2015; 609:58-62. [DOI: 10.1016/j.neulet.2015.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/22/2015] [Accepted: 10/02/2015] [Indexed: 11/30/2022]
|
50
|
Schmidt FM, Nowak C, Kratzsch J, Sander C, Hegerl U, Schönknecht P. Dynamics of melanin-concentrating hormone (MCH) serum levels in major depressive disorder during antidepressant treatment. J Affect Disord 2015; 180:207-13. [PMID: 25932975 DOI: 10.1016/j.jad.2015.03.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND In preclinical studies, the hypothalamic polypeptide melanin-concentrating hormone (MCH) has been shown to be involved in depression-like behavior and modulations of MCH and MCH-receptors were proposed as potential new antidepressant drug targets. METHODS For the first time, MCH serum levels were explored in 30 patients with major depressive disorder (MDD) prior to (T1) and after 2 (T2) and 4 weeks (T3) of antidepressant treatment and in 30 age- and sex-matched healthy controls by applying a fluorescence immunoassay. RESULTS Levels of MCH did not differ significantly between un-medicated patients (444.11±174.63pg/mL SD) and controls (450.68±210.03pg/mL SD). In MDD patients, MCH levels significantly decreased from T1 to T3 (F=4.663; p=0.013). Post-hoc analyses showed that these changes were limited to patients treated with mirtazapine but not escitalopram and female but not male patients. MCH-levels showed high correlations from T1 to T3 (r≥0.964, p<0.001) and were found to correlate significantly with parameters of sleep within the controls. LIMITATIONS Small sample size. No follow-up measures were performed within the control group. CONCLUSIONS Our findings suggest peripheral MCH-levels not to be altered in depression but possibly reflecting depression-related state properties that can be modulated by sleep, medication and sex.
Collapse
Affiliation(s)
- Frank M Schmidt
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany.
| | - Claudia Nowak
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Juergen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany
| | - Christian Sander
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Ulrich Hegerl
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| | - Peter Schönknecht
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany
| |
Collapse
|