1
|
Elberse JD, Saberi A, Ahmadi R, Changizi M, Bi H, Hoffstaedter F, Mander BA, Eickhoff SB, Tahmasian M, Alzheimer’s Disease Neuroimaging Initiative. The interplay between insomnia symptoms and Alzheimer's disease across three main brain networks. Sleep 2024; 47:zsae145. [PMID: 38934787 PMCID: PMC11467060 DOI: 10.1093/sleep/zsae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/20/2024] [Indexed: 06/28/2024] Open
Abstract
STUDY OBJECTIVES Insomnia symptoms are prevalent along the trajectory of Alzheimer's disease (AD), but the neurobiological underpinning of their interaction is poorly understood. Here, we assessed structural and functional brain measures within and between the default mode network (DMN), salience network, and central executive network (CEN). METHODS We selected 320 participants from the ADNI database and divided them by their diagnosis: cognitively normal (CN), Mild Cognitive Impairment (MCI), and AD, with and without self-reported insomnia symptoms. We measured the gray matter volume (GMV), structural covariance (SC), degrees centrality (DC), and functional connectivity (FC), testing the effect and interaction of insomnia symptoms and diagnosis on each index. Subsequently, we performed a within-group linear regression across each network and ROI. Finally, we correlated observed abnormalities with changes in cognitive and affective scores. RESULTS Insomnia symptoms were associated with FC alterations across all groups. The AD group also demonstrated an interaction between insomnia and diagnosis. Within-group analyses revealed that in CN and MCI, insomnia symptoms were characterized by within-network hyperconnectivity, while in AD, within- and between-network hypoconnectivity was ubiquitous. SC and GMV alterations were nonsignificant in the presence of insomnia symptoms, and DC indices only showed network-level alterations in the CEN of AD individuals. Abnormal FC within and between DMN and CEN hubs was additionally associated with reduced cognitive function across all groups, and increased depressive symptoms in AD. CONCLUSIONS We conclude that patients with clinical AD present with a unique pattern of insomnia-related functional alterations, highlighting the profound interaction between both conditions.
Collapse
Affiliation(s)
- Jorik D Elberse
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Amin Saberi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Reihaneh Ahmadi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Faculty of Medicine, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Monir Changizi
- Department of Neurological Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanwen Bi
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Felix Hoffstaedter
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Masoud Tahmasian
- Institute of Neuroscience and Medicine, Brain and Behavior (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Nuclear Medicine, University Hospital and Medical Faculty, University of Cologne, Cologne, Germany
| | | |
Collapse
|
2
|
Lacerda RAV, Desio JAF, Kammers CM, Henkes S, Freitas de Sá M, de Souza EF, da Silva DM, Teixeira Pinheiro Gusmão C, Santos JCCD. Sleep disorders and risk of alzheimer's disease: A two-way road. Ageing Res Rev 2024; 101:102514. [PMID: 39317268 DOI: 10.1016/j.arr.2024.102514] [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: 08/08/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Substantial sleep impairment in patients with Alzheimer's disease (AD) is one of the emerging points for continued efforts to better understand the disease. Individuals without cognitive decline, an important marker of the clinical phase of AD, may show early alterations in the sleep-wake cycle. The objective of this critical narrative review is to explore the bidirectional pathophysiological correlation between sleep disturbances and Alzheimer's Disease. Specifically, it examines how the disruption of sleep homeostasis in individuals without dementia could contribute to the pathogenesis of AD, and conversely, how neurodegeneration in individuals with Alzheimer's Disease might lead to dysregulation of the sleep-wake cycle. Recent scientific results indicate that sleep disturbances, particularly those related to impaired glymphatic clearance, may act as an important mechanism associated with the genesis of Alzheimer's Disease. Additionally, amyloid deposition and tau protein hyperphosphorylation, along with astrocytic hyperactivation, appear to trigger changes in neurotransmission dynamics in areas related to sleep, which may explain the onset of sleep disturbances in individuals with AD. Disruption of sleep homeostasis appears to be a modifiable risk factor in Alzheimer's disease. Whenever possible, the use of non-pharmacological strategies becomes important in this context. From a different perspective, additional research is needed to understand and treat the dysfunction of the sleep-wake cycle in individuals already affected by AD. Early recognition and correction of sleep disturbances in this population could potentially mitigate the progression of dementia and improve the quality of life for those with AD.
Collapse
Affiliation(s)
| | | | | | - Silvana Henkes
- Lutheran University of Brazil - ULBRA, Carazinho, RS, Brazil
| | | | | | | | | | - Júlio César Claudino Dos Santos
- Medical School of the Christus University Center - UNICHRISTUS, Fortaleza, CE, Brazil; Post-Graduate Program of Morphofunctional Sciences, Federal University of Ceara, Fortaleza, CE, Brazil; Unifacvest University Center - UNIFACVEST, Lages, SC, Brazil.
| |
Collapse
|
3
|
Hu Y, He X, Wu Y, Zhang W, Feng H, Liu H, Wu Q, Gao L, Long Y, Li X, Deng J, Ma Y, Li N. Sedative-Hypnotic Effect and Mechanism of Carbon Nanofiber Loaded with Essential Oils of Ligusticum chuanxiong ( Ligusticum chuanxiong Hort.) and Finger Citron ( Citrus medica L. var. sarcodactylis) on Mice Models of Insomnia. Biomolecules 2024; 14:1102. [PMID: 39334868 PMCID: PMC11430208 DOI: 10.3390/biom14091102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
(1) Background: Insomnia is a neurological illness that poses a significant threat to both physical and mental health. It results in the activation of neuroglial cells, heightened neuroinflammation, oxidative stress, and disruptions in the Hypothalamic-Pituitary-Adrenal (HPA) axis. Ligusticum Chuanxiong (CX) and Finger citron (FC) are frequently utilized botanicals for addressing sleeplessness. Both herbs possess notable anti-inflammatory properties in their volatile oils. However, their effectiveness is hindered by the nasal mucosal irritation and instability they exhibit. (2) Methods: This study involved the preparation of a nanofiber composite system using carbon nanofiber (CNF) suspensions containing essential oils of Ligusticum chuanxiong-Finger citron (CXEO-FCEO-CNF). The effects and mechanisms of these essential oils in improving insomnia were investigated using an insomnia mouse model after encapsulation. (3) Results: The CXEO-FCEO-CNF had an average particle size of 103.19 ± 1.64 nm. The encapsulation rates of essential oils of Ligusticum chuanxiong (CXEO) and essential oils of Finger citron (FCEO) were 44.50% and 46.15%, respectively. This resulted in a considerable improvement in the stability of the essential oils over a period of 30 days. The essential oils effectively decreased the irritation of the nasal mucosa following encapsulation. Furthermore, CXEO-FCEO-CNF enhanced voluntary activity and sleep in mice with insomnia, notably boosted the activity of superoxide dismutase (SOD), reduced the concentration of lipoxidized malondialdehyde (MDA), decreased the levels of hormones associated with the HPA axis, and regulated the levels of neurotransmitters, resulting in a beneficial therapeutic outcome. CXEO-FCEO-CNF contains a total of 23 active ingredients, such as alpha-Asarone, (E)-methyl isoeugenol, and Senkyunolide. These ingredients primarily work by modulating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling system to decrease oxidative stress and inflammatory reactions. (4) Conclusions: This study presented initial evidence that the combination of CXEO and FCEO in nanofiber formulations effectively reduces the nasal mucosal irritation and instability of essential oils. Furthermore, it demonstrated the potential anti-neuroinflammatory and therapeutic effects of these formulations in treating insomnia. Overall, this study provides a theoretical foundation for developing new essential oil formulations derived from herbs.
Collapse
Affiliation(s)
- Yue Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiaofang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yuanyuan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Wenjie Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Huiyi Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Haolin Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qianqian Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Leying Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yu Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Xiaoqiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jie Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yin Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
4
|
Brown A, Gervais NJ, Gravelsins L, O'Byrne J, Calvo N, Ramana S, Shao Z, Bernardini M, Jacobson M, Rajah MN, Einstein G. Effects of early midlife ovarian removal on sleep: Polysomnography-measured cortical arousal, homeostatic drive, and spindle characteristics. Horm Behav 2024; 165:105619. [PMID: 39178647 DOI: 10.1016/j.yhbeh.2024.105619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
Bilateral salpingo-oophorectomy (BSO; removal of ovaries and fallopian tubes) prior to age 48 is associated with elevated risk for both Alzheimer's disease (AD) and sleep disorders such as insomnia and sleep apnea. In early midlife, individuals with BSO show reduced hippocampal volume, function, and hippocampal-dependent verbal episodic memory performance associated with changes in sleep. It is unknown whether BSO affects fine-grained sleep measurements (sleep microarchitecture) and how these changes might relate to hippocampal-dependent memory. We recruited thirty-six early midlife participants with BSO. Seventeen of these participants were taking 17β-estradiol therapy (BSO+ET) and 19 had never taken ET (BSO). Twenty age-matched control participants with intact ovaries (AMC) were also included. Overnight at-home polysomnography recordings were collected, along with subjective sleep quality and hot flash frequency. Multivariate Partial Least Squares (PLS) analysis was used to assess how sleep varied between groups. Compared to AMC, BSO without ET was associated with significantly decreased time spent in non-rapid eye movement (NREM) stage 2 sleep as well as increased NREM stage 2 and 3 beta power, NREM stage 2 delta power, and spindle power and maximum amplitude. Increased spindle maximum amplitude was negatively correlated with verbal episodic memory performance. Decreased sleep latency, increased sleep efficiency, and increased time spent in rapid eye movement sleep were observed for BSO+ET. Findings suggest there is an association between ovarian hormone loss and sleep microarchitecture, which may contribute to poorer cognitive outcomes and be ameliorated by ET.
Collapse
Affiliation(s)
- Alana Brown
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada.
| | - Nicole J Gervais
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada; Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9712 CP, the Netherlands.
| | - Laura Gravelsins
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada.
| | - Jordan O'Byrne
- Psychology Department, University of Montreal, Montreal H3T 1J4, Canada; Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal H3G 1M8, Canada.
| | - Noelia Calvo
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada.
| | - Shreeyaa Ramana
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada.
| | - Zhuo Shao
- Genetics Program, North York General Hospital, Toronto M2K 1E1, Canada; Department of Pediatrics, University of Toronto, Toronto M5G 1X8, Canada.
| | | | - Michelle Jacobson
- Princess Margaret Hospital, Toronto M5G 2C4, Canada; Women's College Hospital, Toronto M5S 1B2, Canada.
| | - M Natasha Rajah
- Department of Psychology, Toronto Metropolitan University, Toronto M5B 2K3, Canada.
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto M5S 3G3, Canada; Baycrest Academy of Research and Education, Baycrest Health Sciences, Toronto M6A 2E1, Canada; Tema Genus, Linköping University, Linköping 581 83, Sweden.
| |
Collapse
|
5
|
Ma X, Li J, Yang Y, Qiu X, Sheng J, Han N, Wu C, Xu G, Jiang G, Tian J, Weng X, Wang J. Enhanced cerebral blood flow similarity of the somatomotor network in chronic insomnia: Transcriptomic decoding, gut microbial signatures and phenotypic roles. Neuroimage 2024; 297:120762. [PMID: 39089603 DOI: 10.1016/j.neuroimage.2024.120762] [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: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Chronic insomnia (CI) is a complex disease involving multiple factors including genetics, gut microbiota, and brain structure and function. However, there lacks a unified framework to elucidate how these factors interact in CI. By combining data of clinical assessment, sleep behavior recording, cognitive test, multimodal MRI (structural, functional, and perfusion), gene, and gut microbiota, this study demonstrated that enhanced cerebral blood flow (CBF) similarities of the somatomotor network (SMN) acted as a key mediator to link multiple factors in CI. Specifically, we first demonstrated that only CBF but not morphological or functional networks exhibited alterations in patients with CI, characterized by increases within the SMN and between the SMN and higher-order associative networks. Moreover, these findings were highly reproducible and the CBF similarity method was test-retest reliable. Further, we showed that transcriptional profiles explained 60.4 % variance of the pattern of the increased CBF similarities with the most correlated genes enriched in regulation of cellular and protein localization and material transport, and gut microbiota explained 69.7 % inter-individual variance in the increased CBF similarities with the most contributions from Negativicutes and Lactobacillales. Finally, we found that the increased CBF similarities were correlated with clinical variables, accounted for sleep behaviors and cognitive deficits, and contributed the most to the patient-control classification (accuracy = 84.4 %). Altogether, our findings have important implications for understanding the neuropathology of CI and may inform ways of developing new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Xiaofen Ma
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junle Li
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Yuping Yang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Xiaofan Qiu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Jintao Sheng
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Ningke Han
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Changwen Wu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Guang Xu
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guihua Jiang
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Junzhang Tian
- Department of Nuclear Medicine, Jinan University Affiliated Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xuchu Weng
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China; Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Center for Studies of Psychological Application, South China Normal University, Guangzhou, China; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Jinhui Wang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China; Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China; Center for Studies of Psychological Application, South China Normal University, Guangzhou, China; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
6
|
Chakrabarty M, Chatterjee P, Mukherjee A, Das G, Mollah RI, Mondal B, Sardar S, Basu A, Ghosh M, Sengupta A, Pal SK, Biswas A. Mental health problems raise the odds of cognitive impairment in COVID-19 survivors. Front Psychiatry 2024; 15:1370085. [PMID: 39205850 PMCID: PMC11349739 DOI: 10.3389/fpsyt.2024.1370085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Background COVID-19 survivors around the globe are suffering from mental health issues. While mental health problems can be an early warning sign of dementia, they may also increase the chances of developing the disease. In this study, we examined the mental health of COVID-19 survivors and mapped its associations with cognitive and demographic variables. Method COVID-19 survivors listed in the databases of three tertiary care hospitals in Kolkata were contacted sequentially. 376 willing patients were interviewed over the telephone. 99 COVID-19 patients and 31 matched controls participated in the in-person interviews that were arranged for a more detailed investigation. The participants were administered standardized tests that are widely used for the assessment of cognitive functioning and mental health status. Result 64.89% of COVID-19 survivors reported a deterioration in physical functioning. 44.95% reported a decline in mental health, whereas 41.49% reported a drop in cognitive performance. Detailed investigations revealed that they had an increased risk of having depression, anxiety, and poor sleep quality by 91%, 68%, and 140%, respectively. 6.1% of the patients had mild cognitive impairment, and 4% had dementia. COVID-19 patients who had depression and anxiety were 8.6 and 19.4 times more likely to have cognitive decline, respectively. Compared to the matched controls, COVID-19 patients had greater depression (p<.001), anxiety (p<.001), stress (p =.003), and insomnia (p <.001). They also scored significantly lower on Addenbrooke's Cognitive Examination-III (p =.009) and Picture Naming Test (p =.005) and took significantly longer to complete Trail Making Test-A (p =.002). Conclusion COVID-19 survivors in this study had major mental health issues even one year after contracting the virus. They had significant cognitive deficits that might progress into dementia. Strict monitoring and systematic treatment plans should be implemented as soon as possible.
Collapse
Affiliation(s)
- Madhushree Chakrabarty
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Piali Chatterjee
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Adreesh Mukherjee
- Department of Neuromedicine, North Bengal Medical College, Siliguri, India
| | - Gautam Das
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Rafikul Islam Mollah
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Banshidhar Mondal
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Swarup Sardar
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Ayanendranath Basu
- Interdisciplinary Statistical Research Unit (ISRU), Indian Statistical Institute, Kolkata, India
| | | | - Amitabha Sengupta
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| | - Sankar K. Pal
- Center for Soft Computing Research, A National Facility (CSCR), Indian Statistical Institute, Kolkata, India
| | - Atanu Biswas
- Department of Neuromedicine, Bangur Institute of Neurosciences, Kolkata, India
- Department of Neuromedicine, Institute of Post Graduate Medical Education & Research and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, India
| |
Collapse
|
7
|
Almasi S, Jafarzadeh Shirazi MR, Rezvani MR, Ramezani M, Salehi I, Pegah A, Komaki A. The protective effect of biotin supplementation and swimming training on cognitive impairment and mental symptoms in a rat model of Alzheimer's disease: A behavioral, biochemical, and histological study. Heliyon 2024; 10:e32299. [PMID: 39035497 PMCID: PMC11259780 DOI: 10.1016/j.heliyon.2024.e32299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/14/2024] [Accepted: 05/31/2024] [Indexed: 07/23/2024] Open
Abstract
Vitamin B (Vit B) plays a regulatory role in cognitive memory and learning. We examined the biochemical and behavioral effects of biotin supplementation (BS) and swimming training (ST) on Alzheimer's disease (AD), the most common type of dementia, in male rats. Sixty rats were randomly assigned to six groups: control, sham (receiving phosphate-buffered saline), AD (receiving a single injection of Aβ into the lateral ventricle), ST (for 28 days and before Aβ injection), and BS (receiving BS through oral gavage for 28 days before Aβ injection). The treatments were continued until the end of the behavioral tests. Learning and memory functions were investigated through the Morris water maze (MWM) and depression and anxiety-like behaviors were tested by elevated plus-maze (EPM) and forced swimming tests. In addition, oxidative stress biomarkers, such as total thiol groups (TTG) and malondialdehyde (MDA) in serum were assessed and histological studies were performed using brain tissues. In the AD group, Aβ increased the distance traveled and escape latency in the MWM, but co-administration of BS and ST attenuated the results of the MWM, EPM, and FST tests. Furthermore, BS decreased the litigious biochemical effects of Aβ by enhancing the levels of TTG, in addition to reducing serum MDA levels. The use of BS as a potent antioxidant improved Aβ-induced memory impairment. It attenuated oxidative stress biomarkers in the brain (number of Aβ plaques) and serum of AD rats. We provide evidence for the use of BS in neurodegenerative disorders, such as AD, to elucidate the possible mechanisms.
Collapse
Affiliation(s)
- Shadi Almasi
- Department of Animal Science, School of Agriculture, Shiraz University, Shiraz, Iran
| | | | - Mohammad Reza Rezvani
- Department of Animal Science, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Mahdi Ramezani
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Atefeh Pegah
- Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
8
|
Singhaarachchi PH, Antal P, Calon F, Culmsee C, Delpech JC, Feldotto M, Geertsema J, Hoeksema EE, Korosi A, Layé S, McQualter J, de Rooij SR, Rummel C, Slayo M, Sominsky L, Spencer SJ. Aging, sex, metabolic and life experience factors: Contributions to neuro-inflammaging in Alzheimer's disease research. Neurosci Biobehav Rev 2024; 162:105724. [PMID: 38762130 DOI: 10.1016/j.neubiorev.2024.105724] [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: 02/28/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease (AD) is prevalent around the world, yet our understanding of the disease is still very limited. Recent work suggests that the cornerstone of AD may include the inflammation that accompanies it. Failure of a normal pro-inflammatory immune response to resolve may lead to persistent central inflammation that contributes to unsuccessful clearance of amyloid-beta plaques as they form, neuronal death, and ultimately cognitive decline. Individual metabolic, and dietary (lipid) profiles can differentially regulate this inflammatory process with aging, obesity, poor diet, early life stress and other inflammatory factors contributing to a greater risk of developing AD. Here, we integrate evidence for the interface between these factors, and how they contribute to a pro-inflammatory brain milieu. In particular, we discuss the importance of appropriate polyunsaturated fatty acids (PUFA) in the diet for the metabolism of specialised pro-resolving mediators (SPMs); raising the possibility for dietary strategies to improve AD outlook.
Collapse
Affiliation(s)
| | - Peter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, 1111, Hungary
| | - Frédéric Calon
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec-Laval University, Quebec G1V0A6, Canada; International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, Marburg D-35032, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany
| | - Jean-Christophe Delpech
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Martin Feldotto
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Jorine Geertsema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Emmy E Hoeksema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Sophie Layé
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Jonathan McQualter
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, 1018, the Netherlands
| | - Christoph Rummel
- Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Mary Slayo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Luba Sominsky
- Barwon Health, Geelong, Victoria 3220, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria 3217, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
9
|
Mi W, Meng M, Xu F, Sun L. Efficacy of acupuncture as adjunct therapy for sleep disorders in Parkinson's disease: A systematic review and meta-analysis. Complement Ther Med 2024; 82:103044. [PMID: 38679147 DOI: 10.1016/j.ctim.2024.103044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVE The purpose of this study was to summarize existing clinical studies through a systematic review to explore the efficacy of acupuncture in treating sleep disorders in PD patients. METHODS According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we retrieved the papers through 30 April 2023 from eight databases. The experimental group was treated with acupuncture plus conventional therapy, while the control group was treated with conventional therapy alone or combined with sham acupuncture. The sleep quality was the primary outcome. A team of researchers meticulously performed literature screening, data extraction and risk of bias assessment following the Cochrane Handbook. A meta-analysis was synthesized using Review Manager Version 5.4 software if feasible. The quality of the evidence was assessed by the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) tool. RESULTS A total of 973 papers were identified, with 15 papers involving 957 patients were included in this systematic review. The results showed that acupuncture interventions included manual acupuncture, electroacupuncture, moxibustion and bleeding, with 1-7 times every week implemented during 2-12 weeks. Acupuncture as an adjunct therapy compared to conventional therapy alone showed better effect in sleep quality and overall symptoms of PD. Risk of bias assessment showed deficiencies in blinding and allocation concealment. All included studies were synthesized in a meta-analysis, as the result of which, acupuncture improved PDSS scores(MD =16.57; 95% CI, 7.24-25.90; I2 = 97%) and effective rate for sleep disorders (OR = 5.91; 95% CI, 1.71-20.39; I2 = 54%); meanwhile, acupuncture reduced UPDRS scores(MD = -4.29; 95% CI, -6.54 - -2.03; I2 = 77%) and improved effective rate for PD (OR = 3.22; 95% CI, 1.81-5.72; I2 = 0%). The quality of evidence ranged from low to moderate by GRADE. CONCLUSION This study provides initial evidence that acupuncture as an adjunct therapy might be associated with improvement of sleep disorders in PD. Due to the lack of high-quality studies, larger sample size studies with sham acupuncture groups should be conducted in future. REGISTRATION NUMBER CRD42022364249 (PROSPERO).
Collapse
Affiliation(s)
- Wenjing Mi
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, No.16369 Jingshi Road, Lixia District, Jinan, Shandong 250014, China
| | - Miaomiao Meng
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.16369 Jingshi Road, Lixia District, Jinan, Shandong 250014, China
| | - Fenghua Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.16369 Jingshi Road, Lixia District, Jinan, Shandong 250014, China.
| | - Lingzhi Sun
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.16369 Jingshi Road, Lixia District, Jinan, Shandong 250014, China.
| |
Collapse
|
10
|
Han Z, Yang X, Huang S. Sleep deprivation: A risk factor for the pathogenesis and progression of Alzheimer's disease. Heliyon 2024; 10:e28819. [PMID: 38623196 PMCID: PMC11016624 DOI: 10.1016/j.heliyon.2024.e28819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Sleep deprivation refers to an intentional or unintentional reduction in sleep time, resulting in insufficient sleep. It is often caused by sleep disorders, work demands (e.g., night shifts), and study pressure. Sleep deprivation promotes Aβ deposition and tau hyperphosphorylation, which is a risk factor for the pathogenesis and progression of Alzheimer's disease (AD). Recent research has demonstrated the potential involvement of sleep deprivation in both the pathogenesis and progression of AD through glial cell activation, the glial lymphatic system, orexin system, circadian rhythm system, inflammation, and the gut microbiota. Thus, investigating the molecular mechanisms underlying the association between sleep deprivation and AD is crucial, which may contribute to the development of preventive and therapeutic strategies for AD. This review aims to analyze the impact of sleep deprivation on AD, exploring the underlying pathological mechanisms that link sleep deprivation to the initiation and progression of AD, which offers a theoretical foundation for the development of drugs aimed at preventing and treating AD.
Collapse
Affiliation(s)
- Zhengyun Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingmao Yang
- Ji'nan Zhangqiu District Hospital of Traditional Chinese Medicine, Ji'nan, 250200, China
| | - Shuiqing Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
11
|
Kim RT, Zhou L, Li Y, Krieger AC, Nordvig AS, Butler T, de Leon MJ, Chiang GC. Impaired sleep is associated with tau deposition on 18F-flortaucipir PET and accelerated cognitive decline, accounting for medications that affect sleep. J Neurol Sci 2024; 458:122927. [PMID: 38341949 PMCID: PMC10947806 DOI: 10.1016/j.jns.2024.122927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Impaired sleep is commonly associated with Alzheimer's disease (AD), although the underlying mechanisms remain unclear. Furthermore, the moderating effects of sleep-affecting medications, which have been linked to AD pathology, are incompletely characterized. Using data from the Alzheimer's Disease Neuroimaging Initiative, we investigated whether a medical history of impaired sleep, informant-reported nighttime behaviors, and sleep-affecting medications are associated with beta-amyloid and tau deposition on PET and cognitive change, cross-sectionally and longitudinally. METHODS We included 964 subjects with 18F-florbetapir PET scans. Measures of sleep impairment and medication use were obtained from medical histories and the Neuropsychiatric Inventory Questionnaire. Multivariate models, adjusted for covariates, were used to assess associations among sleep-related features, beta-amyloid and tau, and cognition. Cortical tau deposition, categorized by Braak stage, was assessed using the standardized uptake value peak alignment (SUVP) method on 18F-flortaucipir PET. RESULTS Medical history of sleep impairment was associated with greater baseline tau in the meta-temporal, Braak 1, and Braak 4 regions (p = 0.04, p < 0.001, p = 0.025, respectively). Abnormal nighttime behaviors were also associated with greater baseline tau in the meta-temporal region (p = 0.024), and greater cognitive impairment, cross-sectionally (p = 0.007) and longitudinally (p < 0.001). Impaired sleep was not associated with baseline beta-amyloid (p > 0.05). Short-term use of selective serotonin reuptake inhibitors and benzodiazepines slightly weakened the sleep-tau relationship. CONCLUSIONS Sleep impairment was associated with tauopathy and cognitive decline, which could be linked to increased tau secretion from neuronal hyperactivity. Clinically, our results help identify high-risk individuals who could benefit from sleep-related interventions aimed to delay cognitive decline and AD.
Collapse
Affiliation(s)
- Ryan T Kim
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Bauer-Sherman Fairchild Complex 7 Divinity Avenue, Cambridge, MA 02138, United States of America.
| | - Liangdong Zhou
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Yi Li
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Ana C Krieger
- From the Departments of Medicine and Neurology, Division of Sleep Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 425 E 61st St., 5th Floor, New York, NY 10065, United States of America.
| | - Anna S Nordvig
- From the Department of Neurology, Alzheimer's Disease and Memory Disorders Program, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 428 East 72(nd) Street Suite 500, New York, NY 10021, United States of America.
| | - Tracy Butler
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Mony J de Leon
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Gloria C Chiang
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America; From the Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, United States of America.
| |
Collapse
|
12
|
Aquino G, Benz F, Dressle RJ, Gemignani A, Alfì G, Palagini L, Spiegelhalder K, Riemann D, Feige B. Towards the neurobiology of insomnia: A systematic review of neuroimaging studies. Sleep Med Rev 2024; 73:101878. [PMID: 38056381 DOI: 10.1016/j.smrv.2023.101878] [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: 11/25/2022] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
Insomnia disorder signifies a major public health concern. The development of neuroimaging techniques has permitted to investigate brain mechanisms at a structural and functional level. The present systematic review aims at shedding light on functional, structural, and metabolic substrates of insomnia disorder by integrating the available published neuroimaging data. The databases PubMed, PsycARTICLES, PsycINFO, CINAHL and Web of Science were searched for case-control studies comparing neuroimaging data from insomnia patients and healthy controls. 85 articles were judged as eligible. For every observed finding of each study, the effect size was calculated from standardised mean differences, statistic parameters and figures, showing a marked heterogeneity that precluded a comprehensive quantitative analysis. From a qualitative point of view, considering the findings of significant group differences in the reported regions across the articles, this review highlights the major involvement of the anterior cingulate cortex, thalamus, insula, precuneus and middle frontal gyrus, thus supporting some central themes in the debate on the neurobiology of and offering interesting insights into the psychophysiology of sleep in this disorder.
Collapse
Affiliation(s)
- Giulia Aquino
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine - University of Pisa, Pisa, Italy.
| | - Fee Benz
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Raphael J Dressle
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angelo Gemignani
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine - University of Pisa, Pisa, Italy
| | - Gaspare Alfì
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine - University of Pisa, Pisa, Italy
| | - Laura Palagini
- Department of Experimental and Clinic Medicine, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Kai Spiegelhalder
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dieter Riemann
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernd Feige
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Mattos MK, Zawar I, Manning C, Patrie J, Quigg MS. Interrelationship of Sleep Disturbances and Cognitive Status on Mortality. Am J Alzheimers Dis Other Demen 2024; 39:15333175241252527. [PMID: 38721926 PMCID: PMC11196890 DOI: 10.1177/15333175241252527] [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: 06/27/2024]
Abstract
Sleep disturbances may promote the development and advancement of Alzheimer's disease. Our purpose was to determine if sleep disturbances were associated with earlier mortality while accounting for cognition. The National Alzheimer's Coordinating Center database was used to evaluate mortality risk conferred by sleep, and the Montreal Cognitive Assessment score determined cognitive status. Demographics, sleep disturbances, cognitive status, and comorbid/other neuropsychiatric conditions were examined as predictors of survival time via Cox regression. The sample (N = 31,110) had a median age [interquartile range] of 72 [66, 79] years, MoCA score of 23 [16, 26], and survival time of 106.0 months [104.0,108.0]; 10,278 (33%) died during follow-up; 21% (n = 6461) experienced sleep disturbances. Sleep disturbances impacted survival time depending on cognition, with the greatest effect in transition from normal to cognitive impairment (P < .001). Findings support that sleep disturbances negatively impact survival time, and the impact of sleep disturbances on survival time is interrelated with cognition.
Collapse
Affiliation(s)
- Meghan K. Mattos
- School of Nursing, University of Virginia, Charlottesville, VA 22908, USA
- School of Medicine, Division of Geriatrics, University of Virginia, Charlottesville, VA, USA
| | - Ifrah Zawar
- Comprehensive Epilepsy Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Carol Manning
- Memory Disorders Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - James Patrie
- Division of Biostatistics, Department of Public Health, University of Virginia, Charlottesville, VA, USA
| | - Mark S. Quigg
- Comprehensive Epilepsy Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
14
|
Bozzay ML, Joyce HE, Jiang L, De Vito AN, Emrani S, Browne J, Bayer TA, Quinn MJ, Primack JM, Kelso CM, Wu WC, Rudolph JL, McGeary JE, Kunicki ZJ. Time to Dementia Diagnosis Among Veterans with Comorbid Insomnia and Depressive Episodes. J Alzheimers Dis 2024; 100:899-909. [PMID: 38995783 DOI: 10.3233/jad-240080] [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: 07/14/2024]
Abstract
Background Older adults with heart failure are at elevated risk of Alzheimer's disease and related dementias (AD/ADRD). Research suggests that insomnia and depressive episodes contribute somewhat dissociable impacts on risk for AD/ADRD in this patient population, although the temporal ordering of effects is unknown. Objective This study examined time to dementia diagnosis among patients with comorbid insomnia and/or depressive episodes in an epidemiological sample. Methods Secondary data analyses were conducted using a cohort study of 203,819 Veterans with a primary admission diagnosis of heart failure in 129 VA Medical Centers. Results Patients with diagnoses of both insomnia and depressive episodes had the shortest time to a dementia diagnosis at both 1-year (Hazard ratio = 1.43, 95% CI [1.36, 1.51]) and 3-year follow-up time points (Hazard ratio = 1.40, 95% CI [1.34, 1.47]) versus patients with one or neither comorbidity. Conclusions Individuals with both comorbidities had the shortest time to dementia onset. Screening for these comorbidities may help to identify patients at elevated risk of dementia who could benefit from enhanced monitoring or early intervention strategies for more rapid detection and management of dementia symptoms.
Collapse
Affiliation(s)
- Melanie L Bozzay
- Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Lan Jiang
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
| | - Alyssa N De Vito
- Butler Hospital, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Sheina Emrani
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Julia Browne
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Thomas A Bayer
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - McKenzie J Quinn
- VA RR& D Center for Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, RI, USA
| | - Jennifer M Primack
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Catherine M Kelso
- VA RR& D Center for Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, RI, USA
- Veterans Health Administration, Office of Patient Care Services, Geriatrics and Extended Care, Washington, DC, USA
| | - Wen-Chih Wu
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
| | - James L Rudolph
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - John E McGeary
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Zachary J Kunicki
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
15
|
Weaver DF. Thirty Risk Factors for Alzheimer's Disease Unified by a Common Neuroimmune-Neuroinflammation Mechanism. Brain Sci 2023; 14:41. [PMID: 38248256 PMCID: PMC10813027 DOI: 10.3390/brainsci14010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
One of the major obstacles confronting the formulation of a mechanistic understanding for Alzheimer's disease (AD) is its immense complexity-a complexity that traverses the full structural and phenomenological spectrum, including molecular, macromolecular, cellular, neurological and behavioural processes. This complexity is reflected by the equally complex diversity of risk factors associated with AD. However, more than merely mirroring disease complexity, risk factors also provide fundamental insights into the aetiology and pathogenesis of AD as a neurodegenerative disorder since they are central to disease initiation and subsequent propagation. Based on a systematic literature assessment, this review identified 30 risk factors for AD and then extended the analysis to further identify neuroinflammation as a unifying mechanism present in all 30 risk factors. Although other mechanisms (e.g., vasculopathy, proteopathy) were present in multiple risk factors, dysfunction of the neuroimmune-neuroinflammation axis was uniquely central to all 30 identified risk factors. Though the nature of the neuroinflammatory involvement varied, the activation of microglia and the release of pro-inflammatory cytokines were a common pathway shared by all risk factors. This observation provides further evidence for the importance of immunopathic mechanisms in the aetiopathogenesis of AD.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Research Institute, University Health Network, Departments of Medicine, Chemistry, Pharmaceutical Sciences, University of Toronto, Toronto, ON M5T 0S8, Canada
| |
Collapse
|
16
|
O CK, Siu BWH, Leung VWS, Lin YY, Ding CZ, Lau ESH, Luk AOY, Chow EYK, Ma RCW, Chan JCN, Chan RNY, Wing YK, Kong APS. Association of insomnia with incident chronic cognitive impairment in older adults with type 2 diabetes mellitus: A prospective study of the Hong Kong Diabetes Register. J Diabetes Complications 2023; 37:108598. [PMID: 37716256 DOI: 10.1016/j.jdiacomp.2023.108598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/14/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
AIMS To examine the risk association of insomnia with incident chronic cognitive impairment in older adults with type 2 diabetes mellitus (T2D). METHODS Between July 2010 and June 2015, patients with T2D aged ≥60 years enrolled in the Hong Kong Diabetes Register completed the Insomnia Severity Index (ISI) questionnaire. Patients were considered having insomnia if they had ISI score > 14. We prospectively followed up the cohort and censored outcome through reviewing diagnoses and clinical notes entered by attending physicians in electronic medical record to identify incident cases of mild cognitive impairment and dementia. RESULTS After excluding shift workers and those with established chronic cognitive impairment at baseline, we included 986 patients with T2D in this study (58.3 % men, mean age ± standard deviation: 62.5 ± 2.6 years, disease duration of diabetes: 10.7 ± 8.2 years, HbA1c: 7.4 ± 1.3 %, insulin users: 28.7 %, insomnia: 9.1 %). After a median follow-up of 7.6 (interquartile range = 2.0) years, 41 (4.2 %) developed chronic cognitive impairment. Using Cox regression analysis, insomnia (hazard ratio, HR 2.909, p = 0.012) and HbA1c ≥ 7 % (HR 2.300, p = 0.038) were positively associated with incident chronic cognitive impairment while insulin use (HR 0.309, p = 0.028) showed negative association. CONCLUSIONS Insomnia, suboptimal glycemic control and non-insulin use are independent risk factors for incident chronic cognitive impairment in older adults with T2D.
Collapse
Affiliation(s)
- Chun-Kwan O
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Brian Wai-Hei Siu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Vanessa Wai-Shan Leung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Yuan-Yuan Lin
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Chen-Zhao Ding
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Eric Siu-Him Lau
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Andrea On-Yan Luk
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Elaine Yee-Kwan Chow
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Ronald Ching-Wan Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Juliana Chung-Ngor Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Rachel Ngan-Yin Chan
- Department of Psychiatry, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Yun Kwok Wing
- Department of Psychiatry, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
17
|
Wu G, Wu Y. Neuroprotective effect of Kurarinone against corticosterone-induced cytotoxicity on rat hippocampal neurons by targeting BACE1 to activate P13K-AKT signaling - A potential treatment in insomnia disorder. Pharmacol Res Perspect 2023; 11:e01132. [PMID: 37740616 PMCID: PMC10517343 DOI: 10.1002/prp2.1132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 09/24/2023] Open
Abstract
The hippocampus has been implicated in the pathogenesis of insomnia disorder (ID) and the purpose of this study was to investigate the neuroprotective mechanism of the natural flavone Kurarinone (Kur) on hippocampal neurotoxicity as a potential treatment of ID. The effect of Kur on hippocampal neuronal cell (HNC) viability and apoptosis were assessed by Cell counting kit-8 (CCK-8) assay and flow cytometry, respectively. Then, the effect of Kur on β-site amyloid precursor protein-cleaving enzyme 1 (BACE1), brain-derived neurotrophic factor (BDNF), and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) phosphorylation level were measured by Western blot. Further, SwissTargetPrediction analysis and molecular docking experiments were used to detect a potential target of Kur. Then, the p-chlorophenylalanine (PCPA) model was established in vivo to further study the effect of BACE1 expression on Kur and HNC. As a result, HNC viability was only significantly decreased by 2 μM of Kur. Kur reversed the impacts of corticosterone upon inhibiting viability (0.25-1 μM), PI3K (0.5-1 μM)/AKT phosphorylation, and BDNF (1 μM) level, and enhancing the apoptosis (0.25-1 μM) and BACE1 expression (1 μM) in HNCs. BACE1 was a potential target of Kur. Notably, Kur (150 mg/kg) attenuated PCPA-induced upregulation of BACE1 expression in rat hippocampal tissues as ZRAS (0.8 g/kg). The effects of Kur (1 μM) on corticosterone-treated HNCs were reversed by BACE1 overexpression. Collectively, Kur downregulates BACE1 level to activate PI3K/AKT, thereby attenuating corticosterone-induced toxicity in HNCs, indicating that Kur possibly exerted a neuroprotective effect, which providing a new perspective for the treatment of insomnia disorders.
Collapse
Affiliation(s)
- Guoqing Wu
- Department of MedicineTongde Hospital of Zhejiang ProvinceHangzhouChina
- Zhejiang Institute of Traditional Chinese MedicineHangzhouChina
- Zhejiang Provincial Key Laboratory of New Chinese Medicine Research and DevelopmentHangzhouChina
| | - Yanyan Wu
- Department of MedicineTongde Hospital of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
18
|
Aghelan Z, Pashaee S, Abtahi SH, Karima S, Khazaie H, Ezati M, Khodarahmi R. Natural Immunosuppressants as a Treatment for Chronic Insomnia Targeting the Inflammatory Response Induced by NLRP3/caspase-1/IL-1β Axis Activation: A Scooping Review. J Neuroimmune Pharmacol 2023; 18:294-309. [PMID: 37552452 DOI: 10.1007/s11481-023-10078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/10/2023] [Indexed: 08/09/2023]
Abstract
Chronic insomnia is an inflammatory-related disease with an important pathological basis for various diseases which is a serious threat to a person's physical and mental health. So far, many hypotheses have been proposed to explain the pathogenesis of insomnia, among which inflammatory mechanisms have become the focus of scientific attention. In this regard, the aim of the present scooping review is to evaluate the potential benefits of natural compounds in treatment of chronic insomnia targeting nucleotide-binding oligomerization domain (NOD)-like receptor-pyrin-containing protein 3 (NLRP3)/caspase-1/IL-1β axis as one of the most important activators of inflammatory cascades. The data show that compounds that have the potential to cause inflammation induce sleep disorders, and that inflammatory mediators are key molecules in regulating the sleep-related activity of neurons. In the inflammatory process of insomnia, the role of NLRP3 in the pathogenesis of insomnia has been gradually considered by researchers. NLRP3 is an intracellular sensor that recognizes the widest range of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). After identification and binding to damage factors, NLRP3 inflammasome is assembled to activate the caspase-1 and IL-1β. Increased production and secretion of IL-1β may be involved in central nervous system dysregulation of physiological sleep. The current scooping review reports the potential benefits of natural compounds that target NLRP3 inflammasome pathway activity and highlights the hypothesis which NLRP3 /caspase-1/IL-1β may serve as a potential therapeutic target for managing inflammation and improving symptoms in chronic insomnia.
Collapse
Affiliation(s)
- Zahra Aghelan
- Department of Clinical Biochemistry, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Somayeh Pashaee
- Department of Clinical Biochemistry, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Hosein Abtahi
- Department of Laboratory Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Behehshti University of Medical Sciences, Tehran, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Ezati
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Nurse Street, Kermanshah, 6714415185, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Nurse Street, Kermanshah, 6714415185, Iran.
| |
Collapse
|
19
|
You S, Lv T, Qin R, Hu Z, Ke Z, Yao W, Zhao H, Bai F. Neuro-Navigated rTMS Improves Sleep and Cognitive Impairment via Regulating Sleep-Related Networks' Spontaneous Activity in AD Spectrum Patients. Clin Interv Aging 2023; 18:1333-1349. [PMID: 37601952 PMCID: PMC10439779 DOI: 10.2147/cia.s416992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
Study Objectives By examining spontaneous activity changes of sleep-related networks in patients with the Alzheimer's disease (AD) spectrum with or without insomnia disorder (ID) over time via neuro-navigated repetitive transcranial magnetic stimulation (rTMS), we revealed the effect and mechanism of rTMS targeting the left-angular gyrus in improving the comorbidity symptoms of the AD spectrum with ID. Methods A total of 34 AD spectrum patients were recruited in this study, including 18 patients with ID and the remaining 16 patients without ID. All of them were measured for cognitive function and sleep by using the cognitive and sleep subscales of the neuropsychiatric inventory. The amplitude of low-frequency fluctuation changes in sleep-related networks was revealed before and after neuro-navigated rTMS treatment between these two groups, and the behavioral significance was further explored. Results Affective auditory processing and sensory-motor collaborative sleep-related networks with hypo-spontaneous activity were observed at baseline in the AD spectrum with ID group, while substantial increases in activity were evident at follow-up in these subjects. In addition, longitudinal affective auditory processing, sensory-motor and default mode collaborative sleep-related networks with hyper-spontaneous activity were also revealed at follow-up in the AD spectrum with ID group. In particular, longitudinal changes in sleep-related networks were associated with improvements in sleep quality and episodic memory scores in AD spectrum with ID patients. Conclusion We speculated that left angular gyrus-navigated rTMS therapy may enhance the memory function of AD spectrum patients by regulating the spontaneous activity of sleep-related networks, and it was associated with memory consolidation in the hippocampus-cortical circuit during sleep. Clinical Trial Registration The study was registered at the Chinese Clinical Trial Registry, registration ID: ChiCTR2100050496, China.
Collapse
Affiliation(s)
- Shengqi You
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Tingyu Lv
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Ruomeng Qin
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
| | - Zheqi Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
| | - Zhihong Ke
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
| | - Weina Yao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
- Geriatric Medicine Center, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, People’s Republic of China
| |
Collapse
|
20
|
Wickwire EM, Juday TR, Kelkar M, Heo J, Margiotta C, Frech FH. Economic burden of comorbid insomnia in 5 common medical disease subgroups. J Clin Sleep Med 2023; 19:1293-1302. [PMID: 37394794 PMCID: PMC10315590 DOI: 10.5664/jcsm.10592] [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: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
STUDY OBJECTIVES Approximately 85% of insomnia co-occurs with other disorders. Whereas insomnia was once considered "secondary" to these disorders, it is now widely recognized as an independent condition warranting treatment. While it is clear that insomnia can affect the course of other medical conditions, there is scant literature on the economic impact of comorbid insomnia among patients with common medical conditions. The aim of this study was to determine the economic burden of comorbid insomnia in 5 medical diseases commonly associated with insomnia: type 2 diabetes mellitus (T2DM), cancer undergoing treatment, menopause undergoing hormone replacement therapy, osteoporosis, and Alzheimer's disease and related dementias (ADRDs). METHODS This retrospective cohort study used claims data from the IBM MarketScan Commercial and Medicare Supplemental Databases from January 1, 2014, through December 31, 2019. Insomnia and comorbid disease groups were defined using physician-assigned International Classification of Diseases diagnostic codes. Insomnia medication treatment was defined based on ≥1 prescription fills for the most commonly prescribed insomnia medications (zolpidem, low-dose trazodone, and benzodiazepines [as a class]). For each comorbid disease subgroup, 4 cohorts were created: (1) patients with either treated or untreated insomnia, (2) non-sleep-disordered controls, (3) patients with untreated insomnia, and (4) patients with treated insomnia. RESULTS Sample sizes for individuals with comorbid insomnia ranged from 23,168 (T2DM) to 3,015 (ADRDs). Within each disease subgroup and relative to non-sleep-disordered controls, patients with comorbid insomnia demonstrated greater adjusted health care resource utilization and costs across most points of service. Likewise, relative to individuals with untreated insomnia, those with treated insomnia generally demonstrated greater adjusted health care resource utilization and costs. CONCLUSIONS In this national analysis, both untreated comorbid insomnia and comorbid insomnia treated with commonly prescribed insomnia medications were associated with increased health care resource utilization and costs across most points of service. CITATION Wickwire EM, Juday TR, Kelkar M, Heo J, Margiotta C, Frech FH. Economic burden of comorbid insomnia in 5 common medical disease subgroups. J Clin Sleep Med. 2023;19(7):1293-1302.
Collapse
Affiliation(s)
- Emerson M. Wickwire
- Sleep Disorders Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
21
|
Klimenko MO, Mishchenko TA, Mitaeva YI, Kondakova EV, Mitroshina EV, Vedunova MV. Contribution of Chronic Sleep Deprivation to Age-Related Neurodegeneration in a Mouse Model of Familial Alzheimer's Disease (5xFAD). Neurol Int 2023; 15:778-791. [PMID: 37489355 PMCID: PMC10366916 DOI: 10.3390/neurolint15030049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
Sleep-wake cycle disorders most often accompany the elderly and are frequently associated with the development of neurodegenerative processes, primarily Alzheimer's disease. Sleep disturbances can be diagnosed in patients with AD even before the onset of memory and cognitive impairment, and become more pronounced as the disease progresses. Therefore, the expansion of our knowledge of how sleep relates to AD pathogenesis needs to be addressed as soon as possible. Here, we investigated the influence of chronic sleep deprivation on the motor and orienting-exploratory activity of 5xFAD mice, as well as their spatial learning ability and long-term memory retention. The studies carried out revealed that chronic sleep deprivation negatively affects the processes of spatial memory reconsolidation in 5xFAD mice. This leads to the development of stress-related behavioral responses, including aggressive behavior. In addition, the morphological changes in the cerebral cortex, including changes in the nuclear-cytoplasmic ratio and degradation of neuronal processes are observed. Moreover, we found an increase in the level of total DNA methylation in the blood of the sleep-deprived mice, which may be one of the mechanisms of the two-way relationship between sleep and neurodegeneration.
Collapse
Affiliation(s)
- Maria O Klimenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Tatiana A Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Yaroslava I Mitaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Elena V Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
22
|
Chen C, Wang J, Yang C, Yu H, Zhang B, Yang X, Xiong B, Xie Y, Li S, Zhang Z, Zhu F, Liu J, Liu G, Yang X. Multiomics analysis of human peripheral blood reveals marked molecular profiling changes caused by one night of sleep deprivation. MedComm (Beijing) 2023; 4:e252. [PMID: 37139463 PMCID: PMC10149526 DOI: 10.1002/mco2.252] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/04/2023] [Accepted: 03/12/2023] [Indexed: 05/05/2023] Open
Abstract
Sleep insufficiency is associated with various disorders; the molecular basis is unknown until now. Here, 14 males and 18 females were subjected to short-term (24 h) sleep deprivation, and donated fasting blood samples prior to (day 1) and following (days 2 and 3) short-term sleep deprivation. We used multiple omics techniques to examine changes in volunteers' blood samples that were subjected to integrated, biochemical, transcriptomic, proteomic, and metabolomic analyses. Sleep deprivation caused marked molecular changes (46.4% transcript genes, 59.3% proteins, and 55.6% metabolites) that incompletely reversed by day 3. The immune system in particular neutrophil-mediated processes associated with plasma superoxidase dismutase-1 and S100A8 gene expression was markedly affected. Sleep deprivation decreased melatonin levels and increased immune cells, inflammatory factors and c-reactive protein. By disease enrichment analysis, sleep deprivation induced signaling pathways for schizophrenia and neurodegenerative diseases enriched. In sum, this is the first multiomics approach to show that sleep deprivation causes prominent immune changes in humans, and clearly identified potential immune biomarkers associated with sleep deprivation. This study indicated that the blood profile following sleep disruption, such as may occur among shift workers, may induce immune and central nervous system dysfunction.
Collapse
Affiliation(s)
- Chongyang Chen
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Jing Wang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Chao Yang
- Cognitive Impairment Ward of Neurology DepartmentThe 3rd Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Haitao Yu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bingge Zhang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Bocheng Xiong
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Shupeng Li
- School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio‐cerebrovascular DiseasesJinan University College of PharmacyGuangzhouChina
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology DepartmentThe 3rd Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Jianjun Liu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| | - Gong‐Ping Liu
- Cognitive Impairment Ward of Neurology DepartmentThe 3rd Affiliated Hospital of Shenzhen UniversityShenzhenChina
- Co‐innovation Center of NeurodegenerationNantong UniversityNantongChina
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020‐2024)Shenzhen Center for Disease Control and PreventionShenzhenChina
| |
Collapse
|
23
|
Lin W, Lin YK, Yang FC, Chung CH, Hu JM, Tsao CH, Weng ZX, Ko CA, Chien WC. Risk of neurodegenerative diseases in patients with sleep disorders: A nationwide population-based case-control study. Sleep Med 2023; 107:289-299. [PMID: 37269705 DOI: 10.1016/j.sleep.2023.05.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Our study aimed to explore the associative relationship between neurodegenerative diseases and sleep disorders. PATIENTS This 15-year retrospective longitudinal nationwide population-based matched case-control study used data extracted from the National Health Insurance Research Database. We evaluated 25,589 patients diagnosed with neurodegenerative diseases between 2000 and 2015 and a matched control of 102,356 patients without neurodegenerative diseases. RESULTS Sleep disorders were an independent risk factor for the development of neurodegenerative diseases (adjusted odds ratio (OR): 1.794, 95% confidence interval (CI): 1.235-2.268, P < 0.001), with a positive dose-effect relationship (adjusted OR (95% CI): <1 year: 1.638 (1.093-2.872), P < 0.001; 1-5 years: 1.897 (1.260-3.135), P < 0.001; >5 years: 2.381 (1.467-3.681), P < 0.001. Moreover, patients with sleep disorder and comorbid depression had a significantly higher risk of neurodegenerative disorders (adjusted OR: 5.874). Subgroup analysis showed that insomnia was associated with Alzheimer's disease, Pick's disease and essential tremor (adjusted OR (95% CI): 1.555 (1.069-1.965), 1.934 (1.331-2.445) and 2.089 (1.439-2.648), respectively). Obstructive sleep apnea was associated with Parkinson's disease, essential tremor, and primary dystonia (adjusted OR (95% CI): 1.801 (1.239-2.275), 5.523 (3.802-6.977), and 4.892 (3.365-6.178), respectively). Other specific sleep disorders were associated with Pick's disease, Parkinson's disease, essential tremor, and primary dystonia (adjusted OR (95% CI): 8.901 (6.101-11.010), 1.549 (1.075-1.986), 2.791 (1.924-3.531), and 9.114 (6.283-10.506), respectively). CONCLUSION Sleep disorders are associated with the subsequent development of neurodegenerative disorders. Moreover, sleep disorder patients with comorbid depression have a higher risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Wei Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kai Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
| | - Fu-Chi Yang
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; School of Public Health, National Defense Medical Center, Taipei, Taiwan; Taiwanese Injury Prevention and Safety Promotion Association, Taiwan
| | - Je-Ming Hu
- Division of Colorectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Huei Tsao
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Zi-Xeng Weng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-An Ko
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; School of Public Health, National Defense Medical Center, Taipei, Taiwan; Taiwanese Injury Prevention and Safety Promotion Association, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 114, Taiwan.
| |
Collapse
|
24
|
Masuda K, Katsuda Y, Niwa Y, Sakurai T, Hirano A. Analysis of circadian rhythm components in EEG/EMG data of aged mice. Front Neurosci 2023; 17:1173537. [PMID: 37250413 PMCID: PMC10213445 DOI: 10.3389/fnins.2023.1173537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
Aging disrupts circadian clocks, as evidenced by a reduction in the amplitude of circadian rhythms. Because the circadian clock strongly influences sleep-wake behavior in mammals, age-related alterations in sleep-wake patterns may be attributable, at least partly, to functional changes in the circadian clock. However, the effect of aging on the circadian characteristics of sleep architecture has not been well assessed, as circadian behaviors are usually evaluated through long-term behavioral recording with wheel-running or infrared sensors. In this study, we examined age-related changes in circadian sleep-wake behavior using circadian components extracted from electroencephalography (EEG) and electromyography (EMG) data. EEG and EMG were recorded from 12 to 17-week-old and 78 to 83-week-old mice for 3 days under light/dark and constant dark conditions. We analyzed time-dependent changes in the duration of sleep. Rapid eye movement (REM) and non-REM (NREM) sleep significantly increased during the night phase in old mice, whereas no significant change was observed during the light phase. The circadian components were then extracted from the EEG data for each sleep-wake stage, revealing that the circadian rhythm in the power of delta waves during NREM sleep was attenuated and delayed in old mice. Furthermore, we used machine learning to evaluate the phase of the circadian rhythm, with EEG data serving as the input and the phase of the sleep-wake rhythm (environmental time) as the output. The results indicated that the output time for the old mice data tended to be delayed, specifically at night. These results indicate that the aging process significantly impacts the circadian rhythm in the EEG power spectrum despite the circadian rhythm in the amounts of sleep and wake attenuated but still remaining in old mice. Moreover, EEG/EMG analysis is useful not only for evaluating sleep-wake stages but also for circadian rhythms in the brain.
Collapse
Affiliation(s)
- Kosaku Masuda
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoko Katsuda
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasutaka Niwa
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Medicine, Hirosaki University, Hirosaki, Aomori, Japan
| | - Takeshi Sakurai
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Arisa Hirano
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
25
|
Su T, Calvo RA, Jouaiti M, Daniels S, Kirby P, Dijk DJ, Della Monica C, Vaidyanathan R. Assessing a Sleep Interviewing Chatbot to Improve Subjective and Objective Sleep: Protocol for an Observational Feasibility Study. JMIR Res Protoc 2023; 12:e45752. [PMID: 37166964 DOI: 10.2196/45752] [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: 01/15/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Sleep disorders are common among the aging population and people with neurodegenerative diseases. Sleep disorders have a strong bidirectional relationship with neurodegenerative diseases, where they accelerate and worsen one another. Although one-to-one individual cognitive behavioral interventions (conducted in-person or on the internet) have shown promise for significant improvements in sleep efficiency among adults, many may experience difficulties accessing interventions with sleep specialists, psychiatrists, or psychologists. Therefore, delivering sleep intervention through an automated chatbot platform may be an effective strategy to increase the accessibility and reach of sleep disorder intervention among the aging population and people with neurodegenerative diseases. OBJECTIVE This work aims to (1) determine the feasibility and usability of an automated chatbot (named MotivSleep) that conducts sleep interviews to encourage the aging population to report behaviors that may affect their sleep, followed by providing personalized recommendations for better sleep based on participants' self-reported behaviors; (2) assess the self-reported sleep assessment changes before, during, and after using our automated sleep disturbance intervention chatbot; (3) assess the changes in objective sleep assessment recorded by a sleep tracking device before, during, and after using the automated chatbot MotivSleep. METHODS We will recruit 30 older adult participants from West London for this pilot study. Each participant will have a sleep analyzer installed under their mattress. This contactless sleep monitoring device passively records movements, heart rate, and breathing rate while participants are in bed. In addition, each participant will use our proposed chatbot MotivSleep, accessible on WhatsApp, to describe their sleep and behaviors related to their sleep and receive personalized recommendations for better sleep tailored to their specific reasons for disrupted sleep. We will analyze questionnaire responses before and after the study to assess their perception of our proposed chatbot; questionnaire responses before, during, and after the study to assess their subjective sleep quality changes; and sleep parameters recorded by the sleep analyzer throughout the study to assess their objective sleep quality changes. RESULTS Recruitment will begin in May 2023 through UK Dementia Research Institute Care Research and Technology Centre organized community outreach. Data collection will run from May 2023 until December 2023. We hypothesize that participants will perceive our proposed chatbot as intelligent and trustworthy; we also hypothesize that our proposed chatbot can help improve participants' subjective and objective sleep assessment throughout the study. CONCLUSIONS The MotivSleep automated chatbot has the potential to provide additional care to older adults who wish to improve their sleep in more accessible and less costly ways than conventional face-to-face therapy. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/45752.
Collapse
Affiliation(s)
- Ting Su
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
- Dyson School of Design Engineering, Imperial College London, London, United Kingdom
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
| | - Rafael A Calvo
- Dyson School of Design Engineering, Imperial College London, London, United Kingdom
| | - Melanie Jouaiti
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
| | - Sarah Daniels
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Pippa Kirby
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- Department of Therapies, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Derk-Jan Dijk
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, United Kingdom
| | - Ciro Della Monica
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, United Kingdom
| | - Ravi Vaidyanathan
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
- Care Research & Technology Centre, UK Dementia Research Institute, London, United Kingdom
| |
Collapse
|
26
|
Benca RM, Bertisch SM, Ahuja A, Mandelbaum R, Krystal AD. Wake Up America: National Survey of Patients’ and Physicians’ Views and Attitudes on Insomnia Care. J Clin Med 2023; 12:jcm12072498. [PMID: 37048582 PMCID: PMC10094753 DOI: 10.3390/jcm12072498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
While both patients and physicians consider sleep to be important, sleep health may not receive appropriate consideration during patient visits with health care professionals (HCPs). We completed the first large-scale survey of people with trouble sleeping (PWTS) and physicians who treat insomnia to understand their perspectives and potential discrepancies between them. The Harris Poll conducted online surveys of adult PWTS and HCPs (primary care physicians [PCPs] and psychiatrists) in the United States from September to October 2021. Respondents included 1001 PWTS, 300 PCPs, and 152 psychiatrists. Most HCPs agreed that sleep is critical to good health, yet very few reported routinely conducting full sleep histories on their patients. Approximately 30% of PWTS reported that their PCP never asks about sleep; zero HCPs in this survey reported “never” inquiring. Few HCPs reported being “very satisfied” with current treatment options; 50% of PCPs reported their patients being satisfied. Two-thirds of PWTS did not believe current treatment options adequately improved their sleep. This survey provides evidence that both PWTS and physicians agreed on the importance of sleep, but that treatment is often perceived as ineffective. This survey identifies a need for HCPs to address insomnia management and treatment gaps.
Collapse
|
27
|
Lai Y, Hua L, Yang J, Xu J, Chen J, Zhang S, Zhu S, Li J, Shi S. The Effect of Chinese Agarwood Essential Oil with Cyclodextrin Inclusion against PCPA-Induced Insomnia Rats. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020635. [PMID: 36677694 PMCID: PMC9864866 DOI: 10.3390/molecules28020635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To study the extraction process of agarwood active ingredients (AA) and investigate the safety and effectiveness of AA in the treatment of insomnia rats by nasal administration. METHOD A β-cyclodextrin (β-CD) inclusion compound (a-β-CD) was prepared from agarwood essential oil (AEO), and the preparation process was optimized and characterized. The safety of AA in nasal mucosa was evaluated through Bufo gargarizans maxillary mucosa and rat nasal mucosa models. Insomnia animal models were replicated by injecting p-chlorophenylalanine (PCPA), conducting behavioral tests, and detecting the expression levels of monoamine neurotransmitters (NE and 5-HT) and amino acids (GABA/Glu) in the rat hypothalamus. RESULTS The optimum inclusion process conditions of β-CD were as follows: the feeding ratio was 0.35:1.40 (g:g), the inclusion temperature was 45 °C, the inclusion time was 2 h, and the ICY% and IEO% were 53.78 ± 2.33% and 62.51 ± 3.21%, respectively. The inclusion ratio, temperature, and time are the three factors that have significant effects on the ICY% and IEO% of a-β-CD. AA presented little damage to the nasal mucosa. AA increased the sleep rate, shortened the sleep latency, and prolonged the sleep time of the rats. The behavioral test results showed that AA could ameliorate depression in insomnia rats to a certain extent. The effect on the expression of monoamine neurotransmitters and amino acids in the hypothalamus of rats showed that AA could significantly reduce NE levels and increase the 5-HT level and GABA/Glu ratio in the hypothalamus of insomnia rats. CONCLUSION The preparation of a-β-CD from AEO can reduce its irritation, improve its stability, increase its curative effect, and facilitate its storage and transport. AA have certain therapeutic effects on insomnia. The mechanism of their effect on rat sleep may involve regulating the expression levels of monoamine neurotransmitters and amino acids in the hypothalamus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Senlin Shi
- Correspondence: ; Tel./Fax: +86-13157106148
| |
Collapse
|
28
|
Amyloid-β in Alzheimer's disease - front and centre after all? Neuronal Signal 2023; 7:NS20220086. [PMID: 36687366 PMCID: PMC9829960 DOI: 10.1042/ns20220086] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
The amyloid hypothesis, which proposes that accumulation of the peptide amyloid-β at synapses is the key driver of Alzheimer's disease (AD) pathogenesis, has been the dominant idea in the field of Alzheimer's research for nearly 30 years. Recently, however, serious doubts about its validity have emerged, largely motivated by disappointing results from anti-amyloid therapeutics in clinical trials. As a result, much of the AD research effort has shifted to understanding the roles of a variety of other entities implicated in pathogenesis, such as microglia, astrocytes, apolipoprotein E and several others. All undoubtedly play an important role, but the nature of this has in many cases remained unclear, partly due to their pleiotropic functions. Here, we propose that all of these AD-related entities share at least one overlapping function, which is the local regulation of amyloid-β levels, and that this may be critical to their role in AD pathogenesis. We also review what is currently known of the actions of amyloid-β at the synapse in health and disease, and consider in particular how it might interact with the key AD-associated protein tau in the disease setting. There is much compelling evidence in support of the amyloid hypothesis; rather than detract from this, the implication of many disparate AD-associated cell types, molecules and processes in the regulation of amyloid-β levels may lend further support.
Collapse
|
29
|
Zawar I, Mattos MK, Manning C, Patrie J, Quigg M. Sleep Disturbances Predict Cognitive Decline in Cognitively Healthy Adults. J Alzheimers Dis 2023; 92:1427-1438. [PMID: 36970907 PMCID: PMC10463264 DOI: 10.3233/jad-221244] [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: 04/25/2023]
Abstract
BACKGROUND The effect of nighttime behaviors on cognition has not been studied independently from other neuropsychiatric symptoms. OBJECTIVE We evaluate the following hypotheses that sleep disturbances bring increased risk of earlier cognitive impairment, and more importantly that the effect of sleep disturbances is independent from other neuropsychiatric symptoms that may herald dementia. METHODS We used the National Alzheimer's Coordinating Center database to evaluate the relationship between Neuropsychiatric Inventory Questionnaire (NPI-Q) determined nighttime behaviors which served as surrogate for sleep disturbances and cognitive impairment. Montreal Cognitive Assessment scores defined two groups: conversion from 1) normal to mild cognitive impairment (MCI) and 2) MCI to dementia. The effect of nighttime behaviors at initial visit and covariates of age, sex, education, race, and other neuropsychiatric symptoms (NPI-Q), on conversion risk were analyzed using Cox regression. RESULTS Nighttime behaviors predicted earlier conversion time from normal cognition to MCI (hazard ratio (HR): 1.09; 95% CI: [1.00, 1.48], p = 0.048) but were not associated with MCI to dementia conversion (HR: 1.01; [0.92, 1.10], p = 0.856). In both groups, older age, female sex, lower education, and neuropsychiatric burden increased conversion risk. CONCLUSION Our findings suggest that sleep disturbances predict earlier cognitive decline independently from other neuropsychiatric symptoms that may herald dementia.
Collapse
Affiliation(s)
- Ifrah Zawar
- Department of Neurology, Comprehensive Epilepsy Program, University of Virginia, Charlottesville, VA, USA
| | - Meghan K. Mattos
- School of Nursing, University of Virginia, Charlottesville, VA, USA
- School of Medicine, Division of Geriatrics, University of Virginia, Charlottesville, VA, USA
| | - Carol Manning
- Department of Neurology, Memory Disorders Program, University of Virginia, Charlottesville, VA, USA
| | - James Patrie
- Department of Public Health, Division of Biostatistics, University of Virginia, Charlottesville, VA, USA
| | - Mark Quigg
- Department of Neurology, Comprehensive Epilepsy Program, University of Virginia, Charlottesville, VA, USA
- Sleep Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
30
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin Together with Antibodies to Amyloid Beta Peptide, Phosphorylated Tau, and Tumor Necrosis Factor Alpha Induces Superior Neuroprotection in Alzheimer's Disease Brain Pathology Exacerbated by Sleep Deprivation. ADVANCES IN NEUROBIOLOGY 2023; 32:3-53. [PMID: 37480458 DOI: 10.1007/978-3-031-32997-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Sleep deprivation induces amyloid beta peptide and phosphorylated tau deposits in the brain and cerebrospinal fluid together with altered serotonin metabolism. Thus, it is likely that sleep deprivation is one of the predisposing factors in precipitating Alzheimer's disease (AD) brain pathology. Our previous studies indicate significant brain pathology following sleep deprivation or AD. Keeping these views in consideration in this review, nanodelivery of monoclonal antibodies to amyloid beta peptide (AβP), phosphorylated tau (p-tau), and tumor necrosis factor alpha (TNF-α) in sleep deprivation-induced AD is discussed based on our own investigations. Our results suggest that nanowired delivery of monoclonal antibodies to AβP with p-tau and TNF-α induces superior neuroprotection in AD caused by sleep deprivation, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
31
|
Shukla M, Vincent B. Melatonin as a Harmonizing Factor of Circadian Rhythms, Neuronal Cell Cycle and Neurogenesis: Additional Arguments for Its Therapeutic Use in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:1273-1298. [PMID: 36918783 PMCID: PMC10286584 DOI: 10.2174/1570159x21666230314142505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 03/16/2023] Open
Abstract
The synthesis and release of melatonin in the brain harmonize various physiological functions. The apparent decline in melatonin levels with advanced aging is an aperture to the neurodegenerative processes. It has been indicated that down regulation of melatonin leads to alterations of circadian rhythm components, which further causes a desynchronization of several genes and results in an increased susceptibility to develop neurodegenerative diseases. Additionally, as circadian rhythms and memory are intertwined, such rhythmic disturbances influence memory formation and recall. Besides, cell cycle events exhibit a remarkable oscillatory system, which is downstream of the circadian phenomena. The linkage between the molecular machinery of the cell cycle and complex fundamental regulatory proteins emphasizes the conjectural regulatory role of cell cycle components in neurodegenerative disorders such as Alzheimer's disease. Among the mechanisms intervening long before the signs of the disease appear, the disturbances of the circadian cycle, as well as the alteration of the machinery of the cell cycle and impaired neurogenesis, must hold our interest. Therefore, in the present review, we propose to discuss the underlying mechanisms of action of melatonin in regulating the circadian rhythm, cell cycle components and adult neurogenesis in the context of AD pathogenesis with the view that it might further assist to identify new therapeutic targets.
Collapse
Affiliation(s)
- Mayuri Shukla
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Present Address: Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
32
|
Iacobelli P. Circadian dysregulation and Alzheimer’s disease: A comprehensive review. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Alzheimer’s disease (AD), the foremost variant of dementia, has been associated with a menagerie of risk factors, many of which are considered to be modifiable. Among these modifiable risk factors is circadian rhythm, the chronobiological system that regulates sleep‐wake cycles, food consumption timing, hydration timing, and immune responses amongst many other necessary physiological processes. Circadian rhythm at the level of the suprachiasmatic nucleus (SCN), is tightly regulated in the human body by a host of biomolecular substances, principally the hormones melatonin, cortisol, and serotonin. In addition, photic information projected along afferent pathways to the SCN and peripheral oscillators regulates the synthesis of these hormones and mediates the manner in which they act on the SCN and its substructures. Dysregulation of this cycle, whether induced by environmental changes involving irregular exposure to light, or through endogenous pathology, will have a negative impact on immune system optimization and will heighten the deposition of Aβ and the hyperphosphorylation of the tau protein. Given these correlations, it appears that there is a physiologic association between circadian rhythm dysregulation and AD. This review will explore the physiology of circadian dysregulation in the AD brain, and will propose a basic model for its role in AD‐typical pathology, derived from the literature compiled and referenced throughout.
Collapse
Affiliation(s)
- Peter Iacobelli
- Department of Arts and Sciences, University of South Carolina, Columbia, USA
| |
Collapse
|
33
|
Zhang Q, Zheng L, Luo D, Huang M, Feng Y, Zhao M. Peptide WCPFSRSF alleviates sleep deprivation-induced memory impairment by inhibiting neuroinflammation and modulating IL-6/JAK/STAT signaling pathway. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
34
|
Parrino L, Halasz P, Szucs A, Thomas RJ, Azzi N, Rausa F, Pizzarotti S, Zilioli A, Misirocchi F, Mutti C. Sleep medicine: Practice, challenges and new frontiers. Front Neurol 2022; 13:966659. [PMID: 36313516 PMCID: PMC9616008 DOI: 10.3389/fneur.2022.966659] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep medicine is an ambitious cross-disciplinary challenge, requiring the mutual integration between complementary specialists in order to build a solid framework. Although knowledge in the sleep field is growing impressively thanks to technical and brain imaging support and through detailed clinic-epidemiologic observations, several topics are still dominated by outdated paradigms. In this review we explore the main novelties and gaps in the field of sleep medicine, assess the commonest sleep disturbances, provide advices for routine clinical practice and offer alternative insights and perspectives on the future of sleep research.
Collapse
Affiliation(s)
- Liborio Parrino
- Department of General and Specialized Medicine, Sleep Disorders Center, University Hospital of Parma, Parma, Italy
- *Correspondence: Liborio Parrino
| | - Peter Halasz
- Szentagothai János School of Ph.D Studies, Clinical Neurosciences, Semmelweis University, Budapest, Hungary
| | - Anna Szucs
- Department of Behavioral Sciences, National Institute of Clinical Neurosciences, Semmelweis University, Budapest, Hungary
| | - Robert J. Thomas
- Division of Pulmonary, Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Nicoletta Azzi
- Department of General and Specialized Medicine, Sleep Disorders Center, University Hospital of Parma, Parma, Italy
| | - Francesco Rausa
- Department of General and Specialized Medicine, Sleep Disorders Center, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Silvia Pizzarotti
- Department of General and Specialized Medicine, Sleep Disorders Center, University Hospital of Parma, Parma, Italy
| | - Alessandro Zilioli
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Francesco Misirocchi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Carlotta Mutti
- Department of General and Specialized Medicine, Sleep Disorders Center, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| |
Collapse
|
35
|
Forma F, Pratiwadi R, El-Moustaid F, Smith N, Thorndike F, Velez F. Network meta-analysis comparing the effectiveness of a prescription digital therapeutic for chronic insomnia to medications and face-to-face cognitive behavioral therapy in adults. Curr Med Res Opin 2022; 38:1727-1738. [PMID: 35938209 DOI: 10.1080/03007995.2022.2108616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The purpose of this study was to compare the effectiveness of the only Food and Drug Administration-authorized prescription digital therapeutic (PDT) Somryst versus face-to-face cognitive behavioral therapy for insomnia (CBT-I), or FDA-approved prescription medications for insomnia. METHODS A systematic literature review was undertaken to identify relevant studies. A Bayesian network meta-analysis (NMA) was conducted to examine (1) mean change in insomnia severity index (ISI); (2) proportional change in ISI remitters; (3) mean change in wake after sleep onset (WASO); and (4) mean change in sleep onset latency (SOL). RESULTS Twenty studies provided data on the PDT, CBT-I, CBT-I in combination with self-help (SH), or two prescription medications (eszopiclone and zolpidem). The PDT was associated with significant mean change in ISI (-5.77, 95% Credible Interval [CrI] - 8.53, -3.07) and ISI remitters (OR 12.33; 95% CrI 2.28, 155.91) compared to placebo, and had the highest probability of being the most effective treatment overall for ISI mean change (56%), and ISI remitters (64%). All evaluated interventions significantly outperformed placebo for WASO but no significant differences were observed for SOL (five interventions). Sensitivity analyses excluding medications and meta-regression (assessing type, duration, delivery method for CBT-I) did not affect NMA results. CONCLUSIONS This network meta-analysis demonstrated that a PDT delivering CBT-I had the highest probability of being most effective compared to face-to-face CBT-I, prescription sleep medications, or placebo, as measured by reductions in mean ISI score from baseline and ISI-determined remittance.
Collapse
|
36
|
Study on the potential mechanism, therapeutic drugs and prescriptions of insomnia based on bioinformatics and molecular docking. Comput Biol Med 2022; 149:106001. [DOI: 10.1016/j.compbiomed.2022.106001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/30/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
|
37
|
Page ML, Vance EL, Cloward ME, Ringger E, Dayton L, Ebbert MTW, Miller JB, Kauwe JSK. The Polygenic Risk Score Knowledge Base offers a centralized online repository for calculating and contextualizing polygenic risk scores. Commun Biol 2022; 5:899. [PMID: 36056235 PMCID: PMC9438378 DOI: 10.1038/s42003-022-03795-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/03/2022] [Indexed: 11/20/2022] Open
Abstract
The process of identifying suitable genome-wide association (GWA) studies and formatting the data to calculate multiple polygenic risk scores on a single genome can be laborious. Here, we present a centralized polygenic risk score calculator currently containing over 250,000 genetic variant associations from the NHGRI-EBI GWAS Catalog for users to easily calculate sample-specific polygenic risk scores with comparable results to other available tools. Polygenic risk scores are calculated either online through the Polygenic Risk Score Knowledge Base (PRSKB; https://prs.byu.edu ) or via a command-line interface. We report study-specific polygenic risk scores across the UK Biobank, 1000 Genomes, and the Alzheimer's Disease Neuroimaging Initiative (ADNI), contextualize computed scores, and identify potentially confounding genetic risk factors in ADNI. We introduce a streamlined analysis tool and web interface to calculate and contextualize polygenic risk scores across various studies, which we anticipate will facilitate a wider adaptation of polygenic risk scores in future disease research.
Collapse
Affiliation(s)
- Madeline L Page
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Elizabeth L Vance
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | - Ed Ringger
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Mark T W Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | | | - Justin B Miller
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, USA.,Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
38
|
Aghelan Z, Karima S, Khazaie H, Abtahi SH, Farokhi AR, Rostampour M, Bahrehmand F, Khodarahmi R. IL-1α and TNF-α as an inducer for ROS-mediated NLRP1/NLRP3 inflammasomes activation in mononuclear blood cells from individuals with chronic insomnia disorder. Eur J Neurol 2022; 29:3647-3657. [PMID: 36048129 DOI: 10.1111/ene.15540] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND There are some evidence that cytokines may play an important role in sleep deprivation; however, the underlying mechanisms are still unknown. So, the present study aim to evaluate the relationship between NLRP1 and NLRP3 inflammasomes activation of blood cells and serum levels of cytokines in individuals with chronic insomnia disorder (CID). METHODS Blood samples were collected from 24 individuals with CID and 24 healthy volunteers. The inflammasomes activation was evaluated using real time PCR of NLRP1, NLRP3, ASC, and Caspase-1; western blot of NLRP1 and NLRP3; caspase-1 activity assay; and serum levels of IL-1β, IL-18 and other cytokines using enzyme-linked immunosorbent assay (ELISA). ROS generation in blood cells were detected by flow cytometry assay. As well, MRI scans were obtained on a Siemens Magnetom Avanto 1.5 T MRI whole body scanner using an 8-channel head coil. RESULTS We found the increased activity of NLRP1 and NLRP3 inflammasomes in blood cells; the increased serum levels of pro-inflammatory cytokines; and the decreased serum levels of IL-10 and TGF-β in individuals with CID. We observed significant correlation between increased serum concentration of IL-1β and the severity of insomnia in individuals with CID. The levels of ROS in blood cells was found to be correlated with IL-1α and TNF-α concentrations in serums from individuals with CID. Moreover, the included individuals with CID demonstrated the increased right-cerebellum-cortex and lateral ventricle MD bilaterally compared to controls. CONCLUSIONS This study provided new insights on the pathogenesis of CID and the effects of cytokines on inflammasome activation.
Collapse
Affiliation(s)
- Zahra Aghelan
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Behehshti University of Medical Sciences, Tehran, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Hosein Abtahi
- Department of Laboratory Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Behehshti University of Medical Sciences, Tehran, Iran
| | - Ali Reza Farokhi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoumeh Rostampour
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fariborz Bahrehmand
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
39
|
Forma F, Knight TG, Thorndike FP, Xiong X, Baik R, Velez FF, Maricich YA, Malone DC. Real-World Evaluation of Clinical Response and Long-Term Healthcare Resource Utilization Patterns Following Treatment with a Digital Therapeutic for Chronic Insomnia. CLINICOECONOMICS AND OUTCOMES RESEARCH 2022; 14:537-546. [PMID: 35983014 PMCID: PMC9379126 DOI: 10.2147/ceor.s368780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background and Objectives This analysis evaluated insomnia severity and long-term impact on healthcare resource utilization (HCRU) and costs after treatment with Somryst® (previously called SHUTi), a digital therapeutic delivering cognitive behavioral therapy for insomnia (CBT-I). Methods Change from baseline in insomnia severity index (ISI) score was assessed using last observed ISI score. A pre/post analysis of claims data was conducted, comparing HCRU in patients with self-identified sleep problems who successfully initiated the therapeutic (index date) between June 1, 2016 and December 31, 2018. Results A total of 248 patients were analyzed (median age 56.5 years, 57.3% female, mean ISI score 19.13, 52.4% treated with sleep aid medications pre-index). After 9 weeks, mean ISI score declined by 37.2% from baseline (19.1 vs 12.0), 58.8% of patients achieved ISI responder status (ISI score improved by =>7; NNT: 1.7), and 26.6% of patients achieved insomnia remission (ISI score <8; NNT for remission: 3.8). After two-year follow-up, post-index events were reduced (compared to 2 years pre-index) for emergency department visits (-53%; IRR: 0.47; 95% CI 0.27, 0.82; P=0.008), hospiatizations (-21%; IRR: 0.79; 95% CI 0.46, 1.35; P=0.389) and hospital outpatient visits (-13%; IRR: 0.87; 95% CI 0.66, 1.14; P=0.315). Slightly increased rates were observed for ambulatory surgical center visits (2%; IRR: 1.02; 95% CI 0.73, 1.44; P=0.903) and office visits (2%; IRR: 1.02; 95% CI 0.92, 1.14; P=0.672). The number of patients treated with sleep aid medications dropped 18.5% (52.4% pre-index vs 42.7% post-index). Average number of prescriptions decreased from 3.98 pre-index to 3.73 post-index (P= 0.552). Total two-year cost reduction post-index vs pre-index was $510,678, or -$2059 per patient. Conclusion In a real-world cohort of patients with chronic insomnia, treatment with a digital therapeutic delivering CBT-I was associated with reductions in insomnia severity, emergency department visits, and net costs.
Collapse
Affiliation(s)
- Felicia Forma
- Pear Therapeutics, Inc., Medical Affairs Department, Boston, MA, USA
| | - Tyler G Knight
- Labcorp Drug Development, Market Access Consulting Department, Gaithersburg, MD, USA
| | | | - Xiaorui Xiong
- Pear Therapeutics, Inc., Medical Affairs Department, Boston, MA, USA
| | - Rebecca Baik
- Labcorp Drug Development, Market Access Consulting Department, Gaithersburg, MD, USA
| | - Fulton F Velez
- Pear Therapeutics, Inc., Medical Affairs Department, Boston, MA, USA
| | - Yuri A Maricich
- Pear Therapeutics, Inc., Medical Affairs Department, Boston, MA, USA
| | | |
Collapse
|
40
|
Needham H, Torpey G, Flores CC, Davis CJ, Vanderheyden WM, Gerstner JR. A Dichotomous Role for FABP7 in Sleep and Alzheimer's Disease Pathogenesis: A Hypothesis. Front Neurosci 2022; 16:798994. [PMID: 35844236 PMCID: PMC9280343 DOI: 10.3389/fnins.2022.798994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.
Collapse
Affiliation(s)
- Hope Needham
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Grace Torpey
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - William M. Vanderheyden
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
41
|
Chen D, Wang X, Huang T, Jia J. Sleep and Late-Onset Alzheimer's Disease: Shared Genetic Risk Factors, Drug Targets, Molecular Mechanisms, and Causal Effects. Front Genet 2022; 13:794202. [PMID: 35656316 PMCID: PMC9152224 DOI: 10.3389/fgene.2022.794202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/23/2022] [Indexed: 12/30/2022] Open
Abstract
Late-onset Alzheimer's disease (AD) is associated with sleep-related phenotypes (SRPs). The fact that whether they share a common genetic etiology remains largely unknown. We explored the shared genetics and causality between AD and SRPs by using high-definition likelihood (HDL), cross-phenotype association study (CPASSOC), transcriptome-wide association study (TWAS), and bidirectional Mendelian randomization (MR) in summary-level data for AD (N = 455,258) and summary-level data for seven SRPs (sample size ranges from 359,916 to 1,331,010). AD shared a strong genetic basis with insomnia (r g = 0.20; p = 9.70 × 10-5), snoring (r g = 0.13; p = 2.45 × 10-3), and sleep duration (r g = -0.11; p = 1.18 × 10-3). The CPASSOC identifies 31 independent loci shared between AD and SRPs, including four novel shared loci. Functional analysis and the TWAS showed shared genes were enriched in liver, brain, breast, and heart tissues and highlighted the regulatory roles of immunological disorders, very-low-density lipoprotein particle clearance, triglyceride-rich lipoprotein particle clearance, chylomicron remnant clearance, and positive regulation of T-cell-mediated cytotoxicity pathways. Protein-protein interaction analysis identified three potential drug target genes (APOE, MARK4, and HLA-DRA) that interacted with known FDA-approved drug target genes. The CPASSOC and TWAS demonstrated three regions 11p11.2, 6p22.3, and 16p11.2 may account for the shared basis between AD and sleep duration or snoring. MR showed insomnia had a causal effect on AD (ORIVW = 1.02, P IVW = 6.7 × 10-6), and multivariate MR suggested a potential role of sleep duration and major depression in this association. Our findings provide strong evidence of shared genetics and causation between AD and sleep abnormalities and advance our understanding of the genetic overlap between them. Identifying shared drug targets and molecular pathways can be beneficial for treating AD and sleep disorders more efficiently.
Collapse
Affiliation(s)
- Dongze Chen
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xinpei Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China.,Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China.,Center for Statistical Science, Peking University, Beijing, China
| |
Collapse
|
42
|
Advancement in the contemporary clinical diagnosis and treatment strategies of insomnia disorder. Sleep Med 2022; 91:124-140. [DOI: 10.1016/j.sleep.2022.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/27/2022] [Accepted: 02/19/2022] [Indexed: 11/22/2022]
|
43
|
Clark GT, Yu Y, Urban CA, Fu G, Wang C, Zhang F, Linhardt RJ, Hurley JM. Circadian control of heparan sulfate levels times phagocytosis of amyloid beta aggregates. PLoS Genet 2022; 18:e1009994. [PMID: 35143487 PMCID: PMC8830681 DOI: 10.1371/journal.pgen.1009994] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/14/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's Disease (AD) is a neuroinflammatory disease characterized partly by the inability to clear, and subsequent build-up, of amyloid-beta (Aβ). AD has a bi-directional relationship with circadian disruption (CD) with sleep disturbances starting years before disease onset. However, the molecular mechanism underlying the relationship of CD and AD has not been elucidated. Myeloid-based phagocytosis, a key component in the metabolism of Aβ, is circadianly-regulated, presenting a potential link between CD and AD. In this work, we revealed that the phagocytosis of Aβ42 undergoes a daily circadian oscillation. We found the circadian timing of global heparan sulfate proteoglycan (HSPG) biosynthesis was the molecular timer for the clock-controlled phagocytosis of Aβ and that both HSPG binding and aggregation may play a role in this oscillation. These data highlight that circadian regulation in immune cells may play a role in the intricate relationship between the circadian clock and AD.
Collapse
Affiliation(s)
- Gretchen T. Clark
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
| | - Yanlei Yu
- Rensselaer Polytechnic Institute, Chemistry and Chemical Biology, Troy, New York, United States of America
| | - Cooper A. Urban
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
| | - Guo Fu
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
- Now at the Innovation and Integration Center of New Laser Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chunyu Wang
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
- Rensselaer Polytechnic Institute, Chemistry and Chemical Biology, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Fuming Zhang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Rensselaer Polytechnic Institute, Chemical and Biological Engineering, Troy, New York, United States of America
| | - Robert J. Linhardt
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
- Rensselaer Polytechnic Institute, Chemistry and Chemical Biology, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Rensselaer Polytechnic Institute, Chemical and Biological Engineering, Troy, New York, United States of America
| | - Jennifer M. Hurley
- Rensselaer Polytechnic Institute, Biological Sciences, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| |
Collapse
|
44
|
Zawar I, Mattos MK, Manning C, Quigg M. Sleep Disturbances, Cognitive Status, and Biomarkers of Dementia. J Alzheimers Dis 2022; 89:1367-1374. [PMID: 36031904 PMCID: PMC11218918 DOI: 10.3233/jad-220664] [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: 01/09/2023]
Abstract
BACKGROUND While sleep disturbances appear to be risk factors in Alzheimer's disease (AD) progression, information such as the prevalence across dementia severity and the influence on the trajectory of cognitive decline is unclear. OBJECTIVE We evaluate the hypotheses that the prevalence of insomnia differs by cognitive impairment, that sleep disturbances track with AD biomarkers, and that longitudinal changes in sleep disorders affect cognition. METHODS We used the National Alzheimer's Coordinating Center Database to determine the prevalence of clinician-identified insomnia and nighttime behaviors in normal, mild cognitive impairment (MCI), and demented individuals. We evaluated mean Montreal Cognitive Assessment (MoCA) scores, hippocampal volumes (HV), and CSF phosphorylated tau:amyloid-β ratios at first visit using analysis of variance with age as a covariate. In longitudinal evaluations, we assessed changes in MoCA scores and HV in insomnia and nighttime behaviors between the first and last visits. RESULTS Prevalence of insomnia was 14%, 16%, and 11% for normal, MCI, and dementia groups. Prevalence of nighttime behaviors was 14%, 21%, and 29% respectively. Insomnia patients had higher MoCA scores, larger HV, and lower pTauBeta than individuals without insomnia, indicating less neurodegeneration. In contrast, nighttime behaviors were associated with worse cognition, smaller HV, and higher pTauBeta. Similar findings were seen between longitudinal associations of sleep disorders and cognition and HV. CONCLUSION Our findings suggest that insomnia is unreliably recognized in patients with cognitive impairment. Nighttime behaviors may better indicate the presence of sleep disturbances and have diagnostic specificity in AD over insomnia.
Collapse
Affiliation(s)
- Ifrah Zawar
- Comprehensive Epilepsy Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Meghan K. Mattos
- UVA Section of Geriatrics, School of Nursing, University of Virginia, Charlottesville, VA, USA
| | - Carol Manning
- Memory Disorders Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Mark Quigg
- Comprehensive Epilepsy Program, Department of Neurology, University of Virginia, Charlottesville, VA, USA
- UVA Section of Geriatrics, School of Nursing, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
45
|
Wang C, Yang Y, Ding X, Li J, Zhou X, Teng J, Qi X. Efficacy and safety of Shumian capsules in treating insomnia: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e28194. [PMID: 34918675 PMCID: PMC8678016 DOI: 10.1097/md.0000000000028194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/19/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND It is known to all that the incidence of insomnia is becoming higher and higher with the increase of people's life stress. To some extent, it has brought about bad effects on people's life, work, study, and health, such as mental exhaustion, low work efficiency, and mood irritability. Now there are medications and non-medications methods for insomnia. As one of the treatments for insomnia, western medicine is to prolong the sleeping time and improve the anxious mood. However, taking western medicine to treat insomnia can also be accompanied by some adverse reactions at the same time, such as drug dependence, an allergic reaction, and so on. Traditional Chinese medicine therapy is based on syndrome differentiation and holistic concept. Shumian capsules (SM) are a kind of proprietary Chinese medicine for insomnia, which have the effect of relieving depression and calming the mind. But there are no studies on the efficacy and safety of SM in the treatment of insomnia. Therefore, I will provide a systematic review and meta-analysis to evaluate the efficacy and safety of SM for insomnia. METHODS All the studies searched were from PubMed, EMBASE, Web of Science, Cochrane Library, Chinese National Knowledge Infrastructure, and WanFang databases, and the studies types included in the analysis were all randomized controlled trials. All the retrieval contents were completed independently by 2 researchers, and a third reviewer would be involved when there existed any disagreement. The eligible studies were screened out according to the inclusion criteria and exclusion criteria, and some useful information was extracted and made into a feature table, including the year of the included studies, the age, and disease course of the participants in the studies and intervention methods, etc. Cochrane risk-of-bias tool was used to evaluate the quality of literature and meta-analysis was conducted by RevMan 5.4 software. RESULTS A total of 9 articles including 709 participants were included in the study after screening out. The primary outcomes of statistical analysis were cure rate and total effective rate, while the secondary outcomes included Pittsburgh sleep quality index score and incidence of adverse reactions. The results showed that Pittsburgh sleep quality index score of the SM group and Western medicine group were statistically significant (MD = -0.50, 95% confidence interval [CI] = [-0.78, -0.22], P = .0005). The total effective rate of the SM group was slightly higher than that of the Western medicine group, but there was no statistical significance (relative risk [RR] = 1.03, 95% CI = [0.95,1.13], P = .43). CONCLUSION This meta-analysis provides evidence for the efficacy and safety of SM in the treatment of insomnia, and provides a new idea for the clinical treatment of insomnia. But more research is needed to support further evidence.
Collapse
Affiliation(s)
- Cuiying Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuying Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Ding
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiamin Li
- Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xue Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Teng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
46
|
Aghelan Z, Karima S, Ghadami MR, Khazaie H, Bahrehmand F, Vaisi-Raygani A, Abtahi SH, Khodarahmi R. IgLON5 autoimmunity tested positive in patients with isolated chronic insomnia disease. Clin Exp Immunol 2021; 207:237-240. [PMID: 35020856 PMCID: PMC8982976 DOI: 10.1093/cei/uxab017] [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: 07/11/2021] [Revised: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 02/03/2023] Open
Abstract
In the patients with neurological autoimmune diseases such as anti-IgLON5 disease, insomnia symptoms are very common. Clinical diagnosis of the anti-IgLON5 disease is usually made when neurodegenerative processes have occurred. To find the early signs of anti-IgLON5 disease, we evaluate the presence of IgLON5 autoantibodies in the serum of patients with chronic insomnia disease. Based on video-polysomnography, 22 individuals with isolated chronic insomnia disease were found. A control group of 22 healthy people was chosen using the Pittsburgh Sleep Quality Index (PSQI). An indirect immunofluorescence cell-based test of serum anti-IgLON5 antibodies was used to investigate IgLON5 autoimmunity. Anti-IgLON5 antibodies were detected in the serum of four of these patients with the titer of 1/10. The presence of IgLON5 autoantibodies in some patients with chronic insomnia disease can be considered a causing factor of insomnia which can be effective in more specific treatments of these patients. Moreover, the recognition of anti-IgLON5 disease in the early stages and before the progression of tauopathies can be useful in effective and timely treatment.
Collapse
Affiliation(s)
- Zahra Aghelan
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Behehshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rasoul Ghadami
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran,Habibolah Khazaie, Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Fariborz Bahrehmand
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Asad Vaisi-Raygani
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Hosein Abtahi
- Department of Laboratory Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Behehshti University of Medical Sciences, Tehran, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran,Correspondence: Reza Khodarahmi, Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran. ,
| |
Collapse
|
47
|
Garbarino S, Lanteri P, Bragazzi NL, Magnavita N, Scoditti E. Role of sleep deprivation in immune-related disease risk and outcomes. Commun Biol 2021; 4:1304. [PMID: 34795404 PMCID: PMC8602722 DOI: 10.1038/s42003-021-02825-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Modern societies are experiencing an increasing trend of reduced sleep duration, with nocturnal sleeping time below the recommended ranges for health. Epidemiological and laboratory studies have demonstrated detrimental effects of sleep deprivation on health. Sleep exerts an immune-supportive function, promoting host defense against infection and inflammatory insults. Sleep deprivation has been associated with alterations of innate and adaptive immune parameters, leading to a chronic inflammatory state and an increased risk for infectious/inflammatory pathologies, including cardiometabolic, neoplastic, autoimmune and neurodegenerative diseases. Here, we review recent advancements on the immune responses to sleep deprivation as evidenced by experimental and epidemiological studies, the pathophysiology, and the role for the sleep deprivation-induced immune changes in increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still remain. Further understanding of the causal relationship between sleep deprivation and immune deregulation would help to identify individuals at risk for disease and to prevent adverse health outcomes.
Collapse
Affiliation(s)
- Sergio Garbarino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, 16132, Genoa, Italy.
| | - Paola Lanteri
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, M3J 1P3, Canada
| | - Nicola Magnavita
- Postgraduate School of Occupational Medicine, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Department of Woman/Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100, Lecce, Italy
| |
Collapse
|
48
|
Effect of Acupuncture on Cognitive Function of Insomnia Patients Compared with Drugs: A Protocol for Meta-analysis and Systematic Review. Behav Neurol 2021; 2021:6158275. [PMID: 34552671 PMCID: PMC8452430 DOI: 10.1155/2021/6158275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Insomnia, one of the most common sleep disorders, is thought to have an adverse effect on cognitive function. At the same time, people with cognitive dysfunction are more prone to insomnia. At present, pharmacotherapy is the main treatment for insomnia, but there are some shortcomings such as poor long-term efficacy and potential dependence. There is some evidence that acupuncture has some advantages in alleviating insomnia and improving cognitive function. This study is aimed at investigating the effects of acupuncture and drugs on cognitive function in patients with insomnia and evaluating the efficacy and safety of these two interventions, providing strong evidence for clinical decision-making. The study will retrieve eight major databases: China National Knowledge Infrastructure, Wanfang Database, VIP Database for Chinese Technical Periodicals, SinoMed, PubMed, Web of Science, Embase, and Cochrane Library. Dissertations, conference papers, and ongoing experiments will also be retrieved for supplement. Literature screening and data extraction will be completed by two authors independently (JJ and X-QW). If there were any disagreements, they would be discussed or referred to a third person for adjudication (W-ZW). Authors will use Cochrane risk of bias tool to assess the included studies. The Review Manager Statistical (RevMan) software is used to conduct the statistical process of meta-analysis, and funnel plot is used to evaluate reporting biases. The Grading of Recommendations Assessment Development and Evaluation (GRADE) Profiler can be used to be aware of the quality of evidence.
Collapse
|
49
|
Mattos MK, Chang A, Pitcher K, Whitt C, Ritterband LM, Quigg MS. A Review of Insomnia Treatments for Patients with Mild Cognitive Impairment. Aging Dis 2021; 12:1036-1042. [PMID: 34221547 PMCID: PMC8219491 DOI: 10.14336/ad.2021.0423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
Mild cognitive impairment (MCI) impacts approximately 20% of older adults, with many also experiencing sleep disorders, such as insomnia. Given the relationship between sleep and dementia, addressing sleep issues may offer an opportunity to treat reversible causes. There are two primary treatments for insomnia: behavioral-based (cognitive behavioral therapy for insomnia, CBT-I) and pharmacological interventions. Although CBT-I is recommended as first-line treatment for insomnia in older adults, sedative-hypnotics are more likely to be recommended than non-pharmacological treatments given their convenience and accessibility. However, there are significant concerns in prescribing medications to patients with MCI. To explore this disconnect, we reviewed insomnia treatments in older adults with MCI studies and current guidelines of pharmacological therapy. First, we reviewed studies presenting non-pharmacological treatment of insomnia in older adults with MCI. Although the search yielded over 4,000 non-duplicate titles, only one article presented data on non-pharmacological treatment of insomnia in MCI. The literature covering comorbid insomnia, CBT-I, and MCI is sparse. In contrast to review of non-pharmacological studies, studies on the pharmacological treatment of insomnia in older adults were ample. Finally, we reviewed international guidelines for pharmacological treatment of insomnia in cognitive disorders. More widely used pharmacological interventions show short-term effectiveness with problems of recurrence, ineffectiveness in inadvertent or purposeful chronic use, and adverse side effects. Despite evidence regarding adverse consequences, pharmacological treatment of insomnia remains the most common treatment for insomnia. Reflecting on age-related changes in older adults, particularly those with MCI, inappropriate or mismanagement of medication can lead to unnecessary complications. Further research examining effective behavioral-based sleep management options in older adults with cognitive impairment is needed with exploration of improved sleep on cognitive function.
Collapse
Affiliation(s)
- Meghan K Mattos
- University of Virginia, School of Nursing, Charlottesville, VA 22903, USA.
| | - Angela Chang
- Northwestern University Feinberg School of Medicine, Department of Medical Social Sciences, Chicago, IL 60611, USA.
| | - Katherine Pitcher
- University of Virginia Medical Center, Charlottesville, VA 22903, USA.
| | - Carley Whitt
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Lee M Ritterband
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Mark S Quigg
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| |
Collapse
|
50
|
Poumeaud F, Mircher C, Smith PJ, Faye PA, Sturtz FG. Deciphering the links between psychological stress, depression, and neurocognitive decline in patients with Down syndrome. Neurobiol Stress 2021; 14:100305. [PMID: 33614867 PMCID: PMC7879042 DOI: 10.1016/j.ynstr.2021.100305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/27/2022] Open
Abstract
The relationships between psychological stress and cognitive functions are still to be defined despite some recent progress. Clinically, we noticed that patients with Down syndrome (DS) may develop rapid neurocognitive decline and Alzheimer's disease (AD) earlier than expected, often shortly after a traumatic life event (bereavement over the leave of a primary caregiver, an assault, modification of lifestyle, or the loss of parents). Of course, individuals with DS are naturally prone to develop AD, given the triplication of chromosome 21. However, the relatively weak intensity of the stressful event and the rapid pace of cognitive decline after stress in these patients have to be noticed. It seems DS patients react to stress in a similar manner normal persons react to a very intense stress, and thereafter develop a state very much alike post-traumatic stress disorders. Unfortunately, only a few studies have studied stress-induced regression in patients with DS. Thus, we reviewed the biochemical events involved in psychological stress and found some possible links with cognitive impairment and AD. Interestingly, these links could probably be also applied to non-DS persons submitted to an intense stress. We believe these links should be further explored as a better understanding of the relationships between stress and cognition could help in many situations including individuals of the general population.
Collapse
Affiliation(s)
- François Poumeaud
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
| | - Clotilde Mircher
- Institut Jérôme Lejeune, 37 Rue des Volontaires, F-75015, Paris, France
| | - Peter J. Smith
- University of Chicago, 950 E. 61st Street, SSC Suite 207, Chicago, IL, 60637, USA
| | - Pierre-Antoine Faye
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
- CHU Limoges, Department of Biochemistry and Molecular Biology, F-87000, Limoges, France
| | - Franck G. Sturtz
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
- CHU Limoges, Department of Biochemistry and Molecular Biology, F-87000, Limoges, France
| |
Collapse
|