101
|
Steiger A, Farfan J, Fisher N, Heller HC, Fernandez FX, Ruby NF. Reversible Suppression of Fear Memory Recall by Transient Circadian Arrhythmia. Front Integr Neurosci 2022; 16:900620. [PMID: 35694186 PMCID: PMC9184752 DOI: 10.3389/fnint.2022.900620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
We tested the hypothesis that a temporary period of circadian arrhythmia would transiently impair recall of an aversive memory in Siberian hamsters (Phodopus sungorus). Unlike mice or rats, circadian arrhythmia is easily induced in this species by a one-time manipulation of their ambient lighting [i.e., the disruptive phase shift (DPS) protocol]. Hamsters were conditioned to associate footshocks with a shock chamber (context) and with a predictive auditory tone (cue), and then exposed to the DPS protocol. Following DPS, animals either became arrhythmic (ARR), reentrained to the light-dark cycle (ENT), or became arrhythmic for < 14 days before their circadian locomotor rhythms spontaneously recovered and reentrained (ARR-ENT). Tests for contextual memory showed that freezing was decreased 9–10 days post-DPS when both ARR and ARR-ENT groups were arrhythmic. Once ARR-ENT animals reentrained (day 41), however, freezing was elevated back to Pre-DPS levels and did not differ from those observed in ENT hamsters. ENT animals maintained high levels of freezing at both time points, whereas, freezing remained low in ARR hamsters. In contrast to contextual responses, cued responses were unaffected by circadian arrhythmia; all three groups exhibited elevated levels of freezing in response to the tones. The differential impact of circadian arrhythmia on contextual versus cued associative memory suggests that arrhythmia preferentially impacts memory processes that depend on the hippocampus.
Collapse
Affiliation(s)
- Athreya Steiger
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Julia Farfan
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Nathan Fisher
- Department of Biology, Stanford University, Stanford, CA, United States
| | - H. Craig Heller
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona College of Science, Tucson, AZ, United States
| | - Norman F. Ruby
- Department of Biology, Stanford University, Stanford, CA, United States
- *Correspondence: Norman F. Ruby,
| |
Collapse
|
102
|
Wong H, Buck JM, Borski C, Pafford JT, Keller BN, Milstead RA, Hanson JL, Stitzel JA, Hoeffer CA. RCAN1 knockout and overexpression recapitulate an ensemble of rest-activity and circadian disruptions characteristic of Down syndrome, Alzheimer's disease, and normative aging. J Neurodev Disord 2022; 14:33. [PMID: 35610565 PMCID: PMC9128232 DOI: 10.1186/s11689-022-09444-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Regulator of calcineurin 1 (RCAN1) is overexpressed in Down syndrome (DS), but RCAN1 levels are also increased in Alzheimer's disease (AD) and normal aging. AD is highly comorbid among individuals with DS and is characterized in part by progressive neurodegeneration that resembles accelerated aging. Importantly, abnormal RCAN1 levels have been demonstrated to promote memory deficits and pathophysiology that appear symptomatic of DS, AD, and aging. Anomalous diurnal rest-activity patterns and circadian rhythm disruptions are also common in DS, AD, and aging and have been implicated in facilitating age-related cognitive decline and AD progression. However, no prior studies have assessed whether RCAN1 dysregulation may also promote the age-associated alteration of rest-activity profiles and circadian rhythms, which could in turn contribute to neurodegeneration in DS, AD, and aging. METHODS The present study examined the impacts of RCAN1 deficiency and overexpression on the photic entrainment, circadian periodicity, intensity and distribution, diurnal patterning, and circadian rhythmicity of wheel running in young (3-6 months old) and aged (9-14 months old) mice of both sexes. RESULTS We found that daily RCAN1 levels in the hippocampus and suprachiasmatic nucleus (SCN) of light-entrained young mice are generally constant and that balanced RCAN1 expression is necessary for normal circadian locomotor activity rhythms. While the light-entrained diurnal period was unaltered, RCAN1-null and RCAN1-overexpressing mice displayed lengthened endogenous (free-running) circadian periods like mouse models of AD and aging. In light-entrained young mice, RCAN1 deficiency and overexpression also recapitulated the general hypoactivity, diurnal rest-wake pattern fragmentation, and attenuated amplitudes of circadian activity rhythms reported in DS, preclinical and clinical AD, healthily aging individuals, and rodent models thereof. Under constant darkness, RCAN1-null and RCAN1-overexpressing mice displayed altered locomotor behavior indicating circadian clock dysfunction. Using the Dp(16)1Yey/+ (Dp16) mouse model for DS, which expresses three copies of Rcan1, we found reduced wheel running activity and rhythmicity in both light-entrained and free-running young Dp16 mice like young RCAN1-overexpressing mice. Critically, these diurnal and circadian deficits were rescued in part or entirely by restoring Rcan1 to two copies in Dp16 mice. We also found that RCAN1 deficiency but not RCAN1 overexpression altered protein levels of the clock gene Bmal1 in the SCN. CONCLUSIONS Collectively, this study's findings suggest that both loss and aberrant gain of RCAN1 precipitate anomalous light-entrained diurnal and circadian activity patterns emblematic of DS, AD, and possibly aging.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica T Pafford
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Bailey N Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
| | - Ryan A Milstead
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica L Hanson
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Charles A Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA.
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA.
- Linda Crnic Institute, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
103
|
Yalçin M, Mundorf A, Thiel F, Amatriain-Fernández S, Kalthoff IS, Beucke JC, Budde H, Garthus-Niegel S, Peterburs J, Relógio A. It's About Time: The Circadian Network as Time-Keeper for Cognitive Functioning, Locomotor Activity and Mental Health. Front Physiol 2022; 13:873237. [PMID: 35547585 PMCID: PMC9081535 DOI: 10.3389/fphys.2022.873237] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
A variety of organisms including mammals have evolved a 24h, self-sustained timekeeping machinery known as the circadian clock (biological clock), which enables to anticipate, respond, and adapt to environmental influences such as the daily light and dark cycles. Proper functioning of the clock plays a pivotal role in the temporal regulation of a wide range of cellular, physiological, and behavioural processes. The disruption of circadian rhythms was found to be associated with the onset and progression of several pathologies including sleep and mental disorders, cancer, and neurodegeneration. Thus, the role of the circadian clock in health and disease, and its clinical applications, have gained increasing attention, but the exact mechanisms underlying temporal regulation require further work and the integration of evidence from different research fields. In this review, we address the current knowledge regarding the functioning of molecular circuits as generators of circadian rhythms and the essential role of circadian synchrony in a healthy organism. In particular, we discuss the role of circadian regulation in the context of behaviour and cognitive functioning, delineating how the loss of this tight interplay is linked to pathological development with a focus on mental disorders and neurodegeneration. We further describe emerging new aspects on the link between the circadian clock and physical exercise-induced cognitive functioning, and its current usage as circadian activator with a positive impact in delaying the progression of certain pathologies including neurodegeneration and brain-related disorders. Finally, we discuss recent epidemiological evidence pointing to an important role of the circadian clock in mental health.
Collapse
Affiliation(s)
- Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Freya Thiel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sandra Amatriain-Fernández
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Ida Schulze Kalthoff
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Jan-Carl Beucke
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henning Budde
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Susan Garthus-Niegel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Jutta Peterburs
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
104
|
Yang Y, Zhao JJ, Yu XF. Expert Consensus on Cognitive Dysfunction in Diabetes. Curr Med Sci 2022; 42:286-303. [PMID: 35290601 DOI: 10.1007/s11596-022-2549-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
The incidence of diabetes is gradually increasing in China, and diabetes and associated complications, such as cognitive dysfunction have gained much attention in recent time. However, the concepts, clinical treatment, and prevention of cognitive dysfunction in patients with diabetes remain unclear. The Chinese Society of Endocrinology investigated the current national and overseas situation of cognitive dysfunction associated with diabetes. Based on research both in China and other countries worldwide, the Expert Consensus on Cognitive Dysfunction in Diabetes was established to guide physicians in the comprehensive standardized management of cognitive dysfunction in diabetes and to improve clinical outcomes in Chinese patients. This consensus presents an overview, definition and classification, epidemiology and pathogenesis, risk factors, screening, diagnosis, differential diagnosis, treatment, and prevention of cognitive dysfunction in patients with diabetes.
Collapse
Affiliation(s)
- Yan Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Jun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 25000, China.
| | - Xue-Feng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
105
|
Hanke JM, Schindler KA, Seiler A. On the relationships between epilepsy, sleep, and Alzheimer's disease: A narrative review. Epilepsy Behav 2022; 129:108609. [PMID: 35176650 DOI: 10.1016/j.yebeh.2022.108609] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 12/21/2022]
Abstract
Epilepsy, sleep, and Alzheimer's disease (AD) are tightly and potentially causally interconnected. The aim of our review was to investigate current research directions on these relationships. Our hope is that they may indicate preventive measures and new treatment options for early neurodegeneration. We included articles that assessed all three topics and were published during the last ten years. We found that this literature corroborates connections on various pathophysiological levels, including sleep-stage-related epileptiform activity in AD, the negative consequences of different sleep disorders on epilepsy and cognition, common biochemical pathways as well as network dysfunctions. Here we provide a detailed overview of these topics and we discuss promising diagnostic and therapeutic consequences.
Collapse
Affiliation(s)
- Julie M Hanke
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland
| | - Kaspar A Schindler
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland
| | - Andrea Seiler
- Department of Neurology, Inselspital, Sleep-Wake-Epilepsy-Center, Bern University Hospital, University Bern, Bern, Switzerland.
| |
Collapse
|
106
|
Gębska M, Dalewski B, Pałka Ł, Kołodziej Ł, Sobolewska E. Chronotype Profile, Stress, Depression Level, and Temporomandibular Symptoms in Students with Type D Personality. J Clin Med 2022; 11:jcm11071886. [PMID: 35407492 PMCID: PMC8999628 DOI: 10.3390/jcm11071886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/13/2022] [Accepted: 03/25/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Despite a growing interest in the types of human circadian activity, different chronotypes and personality-related issues have been rarely studied. It has already been emphasized that ‘stress personality’ is considered a risk factor for certain psychosomatic diseases and may be a temporomandibular disorders (TMDs) predictor. Therefore, an attempt has been made to analyze the chronotypes, stress levels, stress factors, and the occurrence of depression and TMDs in students with type D personalities. People with this personality trait tend to experience negative emotions more—depression, anxiety, anger, or hostility—yet may have a negative image of themselves and report somatic complaints. Aim: The aim of this study was to analyze the importance of the chronotype profile for the level of stress perceived, as well as for the occurrence of depression and TMDs in people with type D personalities. Material and Methods: The study has been conducted on a group of 220 physical therapy students. The study group G1 consisted of 110 participants with type D personalities, the control group G2 consisted of the same number of participants without the stress personality. All participants have been analyzed for the chronotype (MEQ), stress perception (PSS10), the occurrence of depression (Beck scale-BDI), the occurrence of TMDs symptoms and have completed the stress factor assessment questionnaire during the study, followed by DS14 questionnaire—a tool for assessing the prevalence of type D personality. Results: In students with type D personalities (G1), the definitely evening and evening chronotypes have been significantly more predominant than in the control group (G2). A significantly higher number of stressors and TMDs symptoms have been observed in the respondents from the G1 group than in the control group (<0.001). Univariate logistic regression analysis showed that type D personality was strongly associated with a more frequent occurrence of all TMD symptoms. Additionally, a significant influence of the evening chronotype on the occurrence of type D personality was observed. Among the potential confounding variables, female gender and a mild and moderate degree of depression have an impact on the occurrence of type D personality (p < 0.05). In the multivariate model, adjusted with the above-mentioned factors, an increased risk of the type D personality trait was found. Conclusion: The evening chronotype and type D personality may imply greater feelings of stress, greater depression, and more frequent symptoms of TMDs in young adults.
Collapse
Affiliation(s)
- Magdalena Gębska
- Department of Rehabilitation Musculoskeletal System, Pomeranian Medical University, 70-204 Szczecin, Poland; (M.G.); (Ł.K.)
| | - Bartosz Dalewski
- Department of Dental Prosthetics, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.D.); (E.S.)
| | - Łukasz Pałka
- Private Dental Practice, 68-200 Zary, Poland
- Correspondence:
| | - Łukasz Kołodziej
- Department of Rehabilitation Musculoskeletal System, Pomeranian Medical University, 70-204 Szczecin, Poland; (M.G.); (Ł.K.)
| | - Ewa Sobolewska
- Department of Dental Prosthetics, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.D.); (E.S.)
| |
Collapse
|
107
|
Xiao Q, Lu J, Zeitzer JM, Matthews CE, Saint-Maurice PF, Bauer C. Rest-activity profiles among U.S. adults in a nationally representative sample: a functional principal component analysis. Int J Behav Nutr Phys Act 2022; 19:32. [PMID: 35331274 PMCID: PMC8944104 DOI: 10.1186/s12966-022-01274-4] [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: 09/15/2021] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Background The 24-h rest and activity behaviors (i.e., physical activity, sedentary behaviors and sleep) are fundamental human behaviors essential to health and well-being. Functional principal component analysis (fPCA) is a flexible approach for characterizing rest-activity rhythms and does not rely on a priori assumptions about the activity shape. The objective of our study is to apply fPCA to a nationally representative sample of American adults to characterize variations in the 24-h rest-activity pattern, determine how the pattern differs according to demographic, socioeconomic and work characteristics, and examine its associations with general health status. Methods The current analysis used data from adults 25 or older in the National Health and Nutrition Examination Survey (NHANES, 2011–2014). Using 7-day 24-h actigraphy recordings, we applied fPCA to derive profiles for overall, weekday and weekend rest-activity patterns. We examined the association between each rest-activity profile in relation to age, gender, race/ethnicity, education, income and working status using multiple linear regression. We also used multiple logistic regression to determine the relationship between each rest-activity profile and the likelihood of reporting poor or fair health. Results We identified four distinct profiles (i.e., high amplitude, early rise, prolonged activity window, biphasic pattern) that together accounted for 86.8% of total variation in the study sample. We identified numerous associations between each rest-activity profile and multiple sociodemographic characteristics. We also found evidence suggesting the associations differed between weekdays and weekends. Finally, we reported that the rest-activity profiles were associated with self-rated health. Conclusions Our study provided evidence suggesting that rest-activity patterns in human populations are shaped by multiple demographic, socioeconomic and work factors, and are correlated with health status. Supplementary Information The online version contains supplementary material available at 10.1186/s12966-022-01274-4.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Epidemiology, Human Genetics and Environmental Health, School of Public Health, the University of Texas Health Science Center at Houston, 1200 Pressler St., TX, Houston, USA.
| | - Jiachen Lu
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, 1200 Pressler St., TX, Houston, USA
| | - Jamie M Zeitzer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Charles E Matthews
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Pedro F Saint-Maurice
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Cici Bauer
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, 1200 Pressler St., TX, Houston, USA.
| |
Collapse
|
108
|
Reynolds C, Mattek N, Lim MM, Beattie Z, Dodge HH, Kaye J. Association Between Mild Cognitive Impairment and Seasonal Rest-Activity Patterns of Older Adults. Front Digit Health 2022; 4:809370. [PMID: 35281221 PMCID: PMC8904352 DOI: 10.3389/fdgth.2022.809370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal variation in rest-activity patterns has been observed in healthy adult populations. This study examined seasonal variation in total time spent overnight in the bedroom by cognitively intact older adults and older adults with mild cognitive impairment (MCI). We hypothesize that seasonal variation in rest-activity patterns is observed in the cognitively intact group and that this variation is disturbed in those with MCI. Study participants were 128 older adults; mean age 85.2 years. Ninety-eight were cognitively intact, and 30 had been diagnosed with MCI. All were enrolled in an ongoing longitudinal study using in-home passive monitoring technology. Infrared presence sensors were placed throughout each participant's home to monitor movement and presence in each room of the home. Activity data was collected from the sensors over a period of up to 527 days. Overnight time in bedroom was found to vary seasonally for the cognitively intact group, with longer times spent overnight in the bedroom during the winter months. This seasonal variation was not observed for those with non-amnestic MCI. MCI is associated with an attenuation of seasonal variation in total time spent in the bedroom at night. Detection of changes in infradian sleep patterns may be an early marker of cognitive decline. Which key determinants are driving these disturbed rhythms, such as features intrinsic to changes in the brain or to environmental factors or external cues, remains an important question for ongoing and future studies.
Collapse
Affiliation(s)
- Christina Reynolds
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: Christina Reynolds
| | - Nora Mattek
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Miranda M. Lim
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Veterans Affairs (VA) Portland Health Care System, Portland, OR, United States
| | - Zachary Beattie
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Hiroko H. Dodge
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Jeffrey Kaye
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| |
Collapse
|
109
|
Wirianto M, Wang CY, Kim E, Koike N, Gomez-Gutierrez R, Nohara K, Escobedo G, Choi JM, Han C, Yagita K, Jung SY, Soto C, Lee HK, Morales R, Yoo SH, Chen Z. The clock modulator Nobiletin mitigates astrogliosis-associated neuroinflammation and disease hallmarks in an Alzheimer's disease model. FASEB J 2022; 36:e22186. [PMID: 35120261 PMCID: PMC8887996 DOI: 10.1096/fj.202101633r] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder, and there is a pressing need to identify disease‐modifying factors and devise interventional strategies. The circadian clock, our intrinsic biological timer, orchestrates various cellular and physiological processes including gene expression, sleep, and neuroinflammation; conversely, circadian dysfunctions are closely associated with and/or contribute to AD hallmarks. We previously reported that the natural compound Nobiletin (NOB) is a clock‐enhancing modulator that promotes physiological health and healthy aging. In the current study, we treated the double transgenic AD model mice, APP/PS1, with NOB‐containing diets. NOB significantly alleviated β‐amyloid burden in both the hippocampus and the cortex, and exhibited a trend to improve cognitive function in these mice. While several systemic parameters for circadian wheel‐running activity, sleep, and metabolism were unchanged, NOB treatment showed a marked effect on the expression of clock and clock‐controlled AD gene expression in the cortex. In accordance, cortical proteomic profiling demonstrated circadian time‐dependent restoration of the protein landscape in APP/PS1 mice treated with NOB. More importantly, we found a potent efficacy of NOB to inhibit proinflammatory cytokine gene expression and inflammasome formation in the cortex, and immunostaining further revealed a specific effect to diminish astrogliosis, but not microgliosis, by NOB in APP/PS1 mice. Together, these results underscore beneficial effects of a clock modulator to mitigate pathological and cognitive hallmarks of AD, and suggest a possible mechanism via suppressing astrogliosis‐associated neuroinflammation.
Collapse
Affiliation(s)
- Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Chih-Yen Wang
- Department of Pediatrics, Baylor College of Medicine, Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ruben Gomez-Gutierrez
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA.,Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain
| | - Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Gabriel Escobedo
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA
| | - Jong Min Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chorong Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Claudio Soto
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA
| | - Hyun Kyoung Lee
- Department of Pediatrics, Baylor College of Medicine, Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA.,Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| |
Collapse
|
110
|
McKee CA, Lee J, Cai Y, Saito T, Saido T, Musiek ES. Astrocytes deficient in circadian clock gene Bmal1 show enhanced activation responses to amyloid-beta pathology without changing plaque burden. Sci Rep 2022; 12:1796. [PMID: 35110643 PMCID: PMC8810760 DOI: 10.1038/s41598-022-05862-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
An emerging link between circadian clock function and neurodegeneration has indicated a critical role for the molecular clock in brain health. We previously reported that deletion of the core circadian clock gene Bmal1 abrogates clock function and induces cell-autonomous astrocyte activation. Regulation of astrocyte activation has important implications for protein aggregation, inflammation, and neuronal survival in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we investigated how astrocyte activation induced by Bmal1 deletion regulates astrocyte gene expression, amyloid-beta (Aβ) plaque-associated activation, and plaque deposition. To address these questions, we crossed astrocyte-specific Bmal1 knockout mice (Aldh1l1-CreERT2;Bmal1fl/fl, termed BMAL1 aKO), to the APP/PS1-21 and the APPNL-G-F models of Aβ accumulation. Transcriptomic profiling showed that BMAL1 aKO induced a unique transcriptional profile affecting genes involved in both the generation and elimination of Aβ. BMAL1 aKO mice showed exacerbated astrocyte activation around Aβ plaques and altered gene expression. However, this astrogliosis did not affect plaque accumulation or neuronal dystrophy in either model. Our results demonstrate that the striking astrocyte activation induced by Bmal1 knockout does not influence Aβ deposition, which indicates that the effect of astrocyte activation on plaque pathology in general is highly dependent on the molecular mechanism of activation.
Collapse
Affiliation(s)
- Celia A McKee
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jiyeon Lee
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yuqi Cai
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Erik S Musiek
- Department of Neurology and Center On Biological Rhythms And Sleep, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
111
|
Chyr J, Gong H, Zhou X. DOTA: Deep Learning Optimal Transport Approach to Advance Drug Repositioning for Alzheimer's Disease. Biomolecules 2022; 12:196. [PMID: 35204697 PMCID: PMC8961573 DOI: 10.3390/biom12020196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States and incurring a substantial global healthcare cost. Unfortunately, current treatments are only palliative and do not cure AD. There is an urgent need to develop novel anti-AD therapies; however, drug discovery is a time-consuming, expensive, and high-risk process. Drug repositioning, on the other hand, is an attractive approach to identify drugs for AD treatment. Thus, we developed a novel deep learning method called DOTA (Drug repositioning approach using Optimal Transport for Alzheimer's disease) to repurpose effective FDA-approved drugs for AD. Specifically, DOTA consists of two major autoencoders: (1) a multi-modal autoencoder to integrate heterogeneous drug information and (2) a Wasserstein variational autoencoder to identify effective AD drugs. Using our approach, we predict that antipsychotic drugs with circadian effects, such as quetiapine, aripiprazole, risperidone, suvorexant, brexpiprazole, olanzapine, and trazadone, will have efficacious effects in AD patients. These drugs target important brain receptors involved in memory, learning, and cognition, including serotonin 5-HT2A, dopamine D2, and orexin receptors. In summary, DOTA repositions promising drugs that target important biological pathways and are predicted to improve patient cognition, circadian rhythms, and AD pathogenesis.
Collapse
Affiliation(s)
- Jacqueline Chyr
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Haoran Gong
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| |
Collapse
|
112
|
Duncan M, Guerriero L, Kohler K, Beechem L, Gillis B, Salisbury F, Wessel C, Wang J, Sunderam S, Bachstetter A, O’Hara B, Murphy M. Chronic Fragmentation of the Daily Sleep-Wake Rhythm Increases Amyloid-beta Levels and Neuroinflammation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Neuroscience 2022; 481:111-122. [PMID: 34856352 PMCID: PMC8941625 DOI: 10.1016/j.neuroscience.2021.11.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/17/2023]
Abstract
Fragmentation of the daily sleep-wake rhythm with increased nighttime awakenings and more daytime naps is correlated with the risk of development of Alzheimer's disease (AD). To explore whether a causal relationship underlies this correlation, the present study tested the hypothesis that chronic fragmentation of the daily sleep-wake rhythm stimulates brain amyloid-beta (Aβ) levels and neuroinflammation in the 3xTg-AD mouse model of AD. Female 3xTg-AD mice were allowed to sleep undisturbed or were subjected to chronic sleep fragmentation consisting of four daily sessions of enforced wakefulness (one hour each) evenly distributed during the light phase, five days a week for four weeks. Piezoelectric sleep recording revealed that sleep fragmentation altered the daily sleep-wake rhythm to resemble the pattern observed in AD. Levels of amyloid-beta (Aβ40 and Aβ42) determined by ELISA were higher in hippocampal tissue collected from sleep-fragmented mice than from undisturbed controls. In contrast, hippocampal levels of tau and phospho-tau differed minimally between sleep fragmented and undisturbed control mice. Sleep fragmentation also stimulated neuroinflammation as shown by increased expression of markers of microglial activation and proinflammatory cytokines measured by q-RT-PCR analysis of hippocampal samples. No significant effects of sleep fragmentation on Aβ, tau, or neuroinflammation were observed in the cerebral cortex. These studies support the concept that improving sleep consolidation in individuals at risk for AD may be beneficial for slowing the onset or progression of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- M.J. Duncan
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| | - L.E. Guerriero
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - K. Kohler
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - L.E. Beechem
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536
| | - B.D. Gillis
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536
| | - F. Salisbury
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - C. Wessel
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - J. Wang
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - S. Sunderam
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - A.D. Bachstetter
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536
| | - B.F. O’Hara
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - M.P. Murphy
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| |
Collapse
|
113
|
A New Perspective on the Treatment of Alzheimer's Disease and Sleep Deprivation-Related Consequences: Can Curcumin Help? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6168199. [PMID: 35069976 PMCID: PMC8769857 DOI: 10.1155/2022/6168199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Sleep disturbances, as well as sleep-wake rhythm disorders, are characteristic symptoms of Alzheimer's disease (AD) that may head the other clinical signs of this neurodegenerative disease. Age-related structural and physiological changes in the brain lead to changes in sleep patterns. Conditions such as AD affect the cerebral cortex, basal forebrain, locus coeruleus, and the hypothalamus, thus changing the sleep-wake cycle. Sleep disorders likewise adversely affect the course of the disease. Since the sleep quality is important for the proper functioning of the memory, impaired sleep is associated with problems in the related areas of the brain that play a key role in learning and memory functions. In addition to synthetic drugs, utilization of medicinal plants has become popular in the treatment of neurological diseases. Curcuminoids, which are in a diarylheptanoid structure, are the main components of turmeric. Amongst them, curcumin has multiple applications in treatment regimens of various diseases such as cardiovascular diseases, obesity, cancer, inflammatory diseases, and aging. Besides, curcumin has been reported to be effective in different types of neurodegenerative diseases. Scientific studies exclusively showed that curcumin leads significant improvements in the pathological process of AD. Yet, its low solubility hence low bioavailability is the main therapeutic limitation of curcumin. Although previous studies have focused different types of advanced nanoformulations of curcumin, new approaches are needed to solve the solubility problem. This review summarizes the available scientific data, as reported by the most recent studies describing the utilization of curcumin in the treatment of AD and sleep deprivation-related consequences.
Collapse
|
114
|
Wanigatunga AA, Liu F, Wang H, Urbanek JK, An Y, Spira AP, Dougherty RJ, Tian Q, Moghekar A, Ferrucci L, Simonsick EM, Resnick SM, Schrack JA. Daily Physical Activity Patterns as a Window on Cognitive Diagnosis in the Baltimore Longitudinal Study of Aging (BLSA). J Alzheimers Dis 2022; 88:459-469. [PMID: 35599480 DOI: 10.3233/jad-215544] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Gradual disengagement from daily physical activity (PA) could signal present or emerging mild cognitive impairment (MCI) or Alzheimer's disease (AD). OBJECTIVE This study examined whether accelerometry-derived patterns of everyday movement differ by cognitive diagnosis in participants of the Baltimore Longitudinal Study of Aging (BLSA). METHODS Activity patterns, overall and by time-of-day, were cross-sectionally compared between participants with adjudicated normal cognition (n = 549) and MCI/AD diagnoses (n = 36; 5 participants [14%] living with AD) using covariate-adjusted regression models. RESULTS Compared to those with normal cognition, those with MCI/AD had 2.1% higher activity fragmentation (SE = 1.0%, p = 0.036) but similar mean total activity counts/day (p = 0.075) and minutes/day spent active (p = 0.174). Time-of-day analyses show MCI/AD participants had lower activity counts and minutes spent active during waking hours (6:00 am-5:59 pm; p < 0.01 for all). Also, they had lower activity fragmentation from 12:00-5:59 am (p < 0.001), but higher fragmentation from 12:00-5:59 pm (p = 0.026). CONCLUSION Differences in the timing and patterns of physical activity throughout the day linked to MCI/AD diagnoses warrant further investigation into potential clinical utility.
Collapse
Affiliation(s)
- Amal A Wanigatunga
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center on Aging and Health, Johns Hopkins University, Baltimore, MD, USA
| | - Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hang Wang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jacek K Urbanek
- Center on Aging and Health, Johns Hopkins University, Baltimore, MD, USA
- Division of Geriatric Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yang An
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Adam P Spira
- Center on Aging and Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ryan J Dougherty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Qu Tian
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Eleanor M Simonsick
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Susan M Resnick
- Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center on Aging and Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
115
|
Atayde AL, Fischer CE, Schweizer TA, Munoz DG. Neuropsychiatric Inventory-Questionnaire Assessed Nighttime Behaviors in Cognitively Asymptomatic Patients with Pathologically Confirmed Alzheimer's Disease Predict More Rapid Cognitive Deterioration. J Alzheimers Dis 2022; 86:1137-1147. [PMID: 35180114 PMCID: PMC9664561 DOI: 10.3233/jad-215276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND The temporal relationship between sleep, Alzheimer's disease (AD), and cognitive impairment remains to be further elucidated. OBJECTIVE First, we aim to determine whether the Neuropsychiatric Inventory-Questionnaire (NPI-Q) assessed nighttime behaviors prior to cognitive decline influence the rate of cognitive deterioration in pathologically confirmed AD, and second, to assess the possible interactions with APOE allele and cerebral amyloid angiopathy (CAA). METHODS The rate of cognitive decline between cognitively asymptomatic participants from the National Alzheimer Coordinating Center who eventually received a neuropathologic diagnosis of AD with (+NTB) or without (-NTB) nighttime behaviors were compared using independent samples t-test. Participants were stratified by APOE carrier and CAA status. Demographic and patient characteristics were assessed using descriptive statistics, and the independent samples t-test was used for continuous variables and chi-square test for categorical variables. The significance level was set at p≤0.05. RESULTS The rate of cognitive decline was greater in +NTB (n = 74; 3.30 points/year) than -NTB (n = 330; 2.45 points/year) (p = 0.016), even if there was no difference in cognitive status at onset. This difference was restricted to APOE ɛ4 carriers (p = 0.049) and positive CAA participants (p = 0.020). Significance was not reached in non-carriers (p = 0.186) and negative CAA (p = 0.364). APOE and CAA were not differentially distributed between the NTB groups. CONCLUSION NPI-Q assessed nighttime behaviors, a surrogate for sleep disturbances, are associated with more rapidly deteriorating cognition in patients with AD neuropathology who are also carriers of APOE ɛ4 or show CAA.
Collapse
Affiliation(s)
- Adrienne L. Atayde
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
| | - Corinne E. Fischer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Faculty of Medicine, Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Tom A. Schweizer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Canada
- Division of Neurosurgery, St. Michael’s Hospital, Toronto, Canada
| | - David G. Munoz
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Pathology, St. Michael’s Hospital, Toronto, Canada
| |
Collapse
|
116
|
Caliandro R, Streng AA, van Kerkhof LWM, van der Horst GTJ, Chaves I. Social Jetlag and Related Risks for Human Health: A Timely Review. Nutrients 2021; 13:nu13124543. [PMID: 34960096 PMCID: PMC8707256 DOI: 10.3390/nu13124543] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/11/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
The term social jetlag is used to describe the discrepancy between biological time, determined by our internal body clock, and social times, mainly dictated by social obligations such as school or work. In industrialized countries, two-thirds of the studying/working population experiences social jetlag, often for several years. Described for the first time in 2006, a considerable effort has been put into understanding the effects of social jetlag on human physiopathology, yet our understanding of this phenomenon is still very limited. Due to its high prevalence, social jetlag is becoming a primary concern for public health. This review summarizes current knowledge regarding social jetlag, social jetlag associated behavior (e.g., unhealthy eating patterns) and related risks for human health.
Collapse
Affiliation(s)
- Rocco Caliandro
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (R.C.); (A.A.S.); (G.T.J.v.d.H.)
| | - Astrid A. Streng
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (R.C.); (A.A.S.); (G.T.J.v.d.H.)
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands;
| | - Linda W. M. van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands;
| | - Gijsbertus T. J. van der Horst
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (R.C.); (A.A.S.); (G.T.J.v.d.H.)
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (R.C.); (A.A.S.); (G.T.J.v.d.H.)
- Correspondence: ; Tel.: +31-10-704-3456; Fax: +31-10-704-4743
| |
Collapse
|
117
|
Bacalini MG, Palombo F, Garagnani P, Giuliani C, Fiorini C, Caporali L, Stanzani Maserati M, Capellari S, Romagnoli M, De Fanti S, Benussi L, Binetti G, Ghidoni R, Galimberti D, Scarpini E, Arcaro M, Bonanni E, Siciliano G, Maestri M, Guarnieri B, Martucci M, Monti D, Carelli V, Franceschi C, La Morgia C, Santoro A. Association of rs3027178 polymorphism in the circadian clock gene PER1 with susceptibility to Alzheimer's disease and longevity in an Italian population. GeroScience 2021; 44:881-896. [PMID: 34921659 PMCID: PMC9135916 DOI: 10.1007/s11357-021-00477-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/15/2021] [Indexed: 12/11/2022] Open
Abstract
Many physiological processes in the human body follow a 24-h circadian rhythm controlled by the circadian clock system. Light, sensed by retina, is the predominant “zeitgeber” able to synchronize the circadian rhythms to the light-dark cycles. Circadian rhythm dysfunction and sleep disorders have been associated with aging and neurodegenerative diseases including mild cognitive impairment (MCI) and Alzheimer’s disease (AD). In the present study, we aimed at investigating the genetic variability of clock genes in AD patients compared to healthy controls from Italy. We also included a group of Italian centenarians, considered as super-controls in association studies given their extreme phenotype of successful aging. We analyzed the exon sequences of eighty-four genes related to circadian rhythms, and the most significant variants identified in this first discovery phase were further assessed in a larger independent cohort of AD patients by matrix assisted laser desorption/ionization-time of flight mass spectrometry. The results identified a significant association between the rs3027178 polymorphism in the PER1 circadian gene with AD, the G allele being protective for AD. Interestingly, rs3027178 showed similar genotypic frequencies among AD patients and centenarians. These results collectively underline the relevance of circadian dysfunction in the predisposition to AD and contribute to the discussion on the role of the relationship between the genetics of age-related diseases and of longevity.
Collapse
Affiliation(s)
- Maria Giulia Bacalini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Laboratorio Brain Aging, Bologna, Italy
| | - Flavia Palombo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy.,CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy.,Department of Laboratory Medicine, Clinical Chemistry, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Cristina Giuliani
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.,Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudio Fiorini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Leonardo Caporali
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | | | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Martina Romagnoli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Sara De Fanti
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.,Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Luisa Benussi
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy
| | - Enrica Bonanni
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelangelo Maestri
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Biancamaria Guarnieri
- Center of Sleep Medicine, Villa Serena Hospital and Villaserena Foundation for the Research, Città S. Angelo, Pescara, Italy
| | | | - Morena Martucci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Department of Applied Mathematics, Institute of Information Technology, Mathematics and Mechanics (ITMM), Lobachevsky State University of Nizhny Novgorod-National Research University (UNN), Nizhny Novgorod, Russia
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy. .,Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.
| |
Collapse
|
118
|
Smagula SF, Stahl ST, Krafty RT, Buysse DJ. Initial proof of concept that a consumer wearable can be used for real-time rest-activity rhythm monitoring. Sleep 2021; 45:6472395. [PMID: 34931683 DOI: 10.1093/sleep/zsab288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Stephen F Smagula
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah T Stahl
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert T Krafty
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Daniel J Buysse
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
119
|
Amyloid Burden in Alzheimer's Disease Patients Is Associated with Alterations in Circadian Rhythm. Dement Neurocogn Disord 2021; 20:99-107. [PMID: 34795773 PMCID: PMC8585536 DOI: 10.12779/dnd.2021.20.4.99] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose In this study we evaluated the relationship between amyloid-beta (Aβ) deposition and 3 aspects of sleep quality in a group of clinically diagnosed Alzheimer's disease (AD) patients. Methods We used self-report questionnaires to assess the quality of sleep using 3 previously established surveys: the Glasgow Sleep Effort Scale (GSES), the Pittsburgh Sleep Quality Index (PSQI), and the Morningness-Eveningness Questionnaire (MEQ). These questionnaires focused on the sleep effort, sleep efficiency, and circadian rhythm patterns of each participant. Also, we evaluated the regional distribution of Aβ in the brain by amyloid positron emission tomography-computed tomography (PET-CT) standardized uptake value ratios (SUVRs) in healthy normal (HN), mild cognitive impairment (MCI), and AD dementia groups. The MCI and AD dementia groups were combined to form the group with cognitive impairment due to AD (CIAD). Results GSES and MEQ scores differed significantly between the HN, MCI, and AD dementia groups (p<0.037), whereas PSQI scores were similar across the groups (p=0.129). GSES and MEQ scores also differed between the HN and CIAD groups (p<0.018). Circadian rhythm scores positively correlated with amyloid PET-CT SUVR in posterior cingulate cortices (p<0.049). Conclusions Sleep effort and abnormal shifts in circadian rhythm were more significant in the CIAD group than in the HN group. At the same time, HN subjects had minimal sleep disturbance, irrespective of clinical status. Thus, alterations in circadian rhythm may be indicative of neurodegeneration due to Aβ deposition.
Collapse
|
120
|
Gao F, Liu T, Tuo M, Chi S. The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target. Neurosci Lett 2021; 765:136247. [PMID: 34530113 DOI: 10.1016/j.neulet.2021.136247] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/01/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
Accumulating evidence has shown that sleep disturbance is a common symptom in Alzheimer's disease (AD), which is regarded as a modifiable risk factor for AD. Orexin is a key modulator of the sleep-wake cycle and has been found to be dysregulated in AD patients. The increased orexin in cerebrospinal fluid (CSF) is associated with decreased sleep efficiency and REM sleep, as well as cognitive impairment in AD patients. The orexin system has profuse projections to brain regions that are implicated in arousal and cognition and has been found to participate in the progression of AD pathology. Conversely the orexin receptor antagonists are able to consolidate sleep and reduce AD pathology. Therefore, improved understanding of the mechanisms linking orexin system, sleep disturbance and AD could make orexin receptor antagonists a promising target for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Fan Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miao Tuo
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
121
|
Abstract
Endogenous biological clocks, orchestrated by the suprachiasmatic nucleus, time the circadian rhythms that synchronize physiological and behavioural functions in humans. The circadian system influences most physiological processes, including sleep, alertness and cognitive performance. Disruption of circadian homeostasis has deleterious effects on human health. Neurodegenerative disorders involve a wide range of symptoms, many of which exhibit diurnal variations in frequency and intensity. These disorders also disrupt circadian homeostasis, which in turn has negative effects on symptoms and quality of life. Emerging evidence points to a bidirectional relationship between circadian homeostasis and neurodegeneration, suggesting that circadian function might have an important role in the progression of neurodegenerative disorders. Therefore, the circadian system has become an attractive target for research and clinical care innovations. Studying circadian disruption in neurodegenerative disorders could expand our understanding of the pathophysiology of neurodegeneration and facilitate the development of novel, circadian-based interventions for these disabling disorders. In this Review, we discuss the alterations to the circadian system that occur in movement (Parkinson disease and Huntington disease) and cognitive (Alzheimer disease and frontotemporal dementia) neurodegenerative disorders and provide directions for future investigations in this field.
Collapse
|
122
|
Posner AB, Tranah GJ, Blackwell T, Yaffe K, Ancoli-Israel S, Redline S, Leng Y, Zeitzer JM, Chen DM, Webber KR, Stone KL. Predicting incident dementia and mild cognitive impairment in older women with nonparametric analysis of circadian activity rhythms in the Study of Osteoporotic Fractures. Sleep 2021; 44:zsab119. [PMID: 33964167 PMCID: PMC8503832 DOI: 10.1093/sleep/zsab119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/31/2021] [Indexed: 11/12/2022] Open
Abstract
STUDY OBJECTIVES Disrupted daily rhythms are associated with mild cognitive impairment (MCI) and dementia. The specific nature of how rhythms and cognition are related, however, is unknown. We hypothesized characteristics from a nonparametric estimate of circadian rest-activity rhythm patterns would be associated to the development of MCI or dementia. METHODS Wrist actigraphy from 1232 cognitively healthy, community-dwelling women (mean age 82.6 years) from the Study of Osteoporotic Fractures was used to estimate rest-activity patterns, including intradaily variability (IV), interdaily stability (IS), most active 10-hour period (M10), least active 5-hour period (L5), and relative amplitude (RA). Logistic regression examined associations of these predictors with 5-year incidence of MCI or dementia. Models were adjusted for potential confounders. RESULTS Women with earlier sleep/wake times had higher risk of dementia, but not MCI, (early vs. average L5 midpoint: OR, 1.66; 95% CI, 1.08-2.55) as did women with smaller day/night activity differentials (low vs. high RA: OR, 1.96; 95% CI, 1.14-3.35). IV, IS, and M10 were not associated with MCI or dementia. CONCLUSION The timing and difference in day/night amplitude, but not variability of activity, may be useful as predictors of dementia.
Collapse
Affiliation(s)
- Alexander B Posner
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
- Division of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Gregory J Tranah
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - Terri Blackwell
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - Kristine Yaffe
- San Francisco VA Medical Center, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Sonia Ancoli-Israel
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Susan Redline
- Departments of Medicine and Neurology, Brigham and Women’s Hospital, and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yue Leng
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jamie M Zeitzer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA
- Mental Illness Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Stanford Center for Sleep Sciences and Medicine, Stanford University, Palo Alto, CA, USA
| | - Dorothy M Chen
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
- Division of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Katey R Webber
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
- Division of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Katie L Stone
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| |
Collapse
|
123
|
Targa ADS, Benítez ID, Dakterzada F, Fontenele-Araujo J, Minguez O, Zetterberg H, Blennow K, Barbé F, Piñol-Ripoll G. The circadian rest-activity pattern predicts cognitive decline among mild-moderate Alzheimer's disease patients. Alzheimers Res Ther 2021; 13:161. [PMID: 34563258 PMCID: PMC8466995 DOI: 10.1186/s13195-021-00903-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/12/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Alterations in circadian rhythms are present in the presymptomatic stage of Alzheimer's disease (AD), possibly contributing to its pathogenesis. However, it is unknown whether such alterations are associated with worse outcomes once individuals are diagnosed with symptomatic disease. We aimed to evaluate the association between the circadian rest-activity pattern and AD-related features in patients with mild-moderate AD. METHODS We assessed the circadian rest-activity pattern of consecutive patients with mild-moderate AD through actigraphy for 14 days. Cerebrospinal fluid was obtained to determine the levels of important pathological markers including amyloid-beta protein (Aβ42), phosphorylated tau (P-tau), total tau (T-tau), and neurofilament light (NF-L). Neuropsychological evaluation was conducted at the beginning of the study and after 12 months of follow-up. Linear regression models were performed considering the global population and Aβ42+ patients only. RESULTS The cohort included 100 patients with mild-moderate AD. The median age [p25;p75] was 76.0 [73.0;80.0] years and 63.0% were female. Older age (effect size [SE] of 0.324 [0.096]; p = 0.001) and male sex (0.780 [0.193]; p = 0.001) were associated with increased fragmentation and decreased synchronization of the rhythm, respectively. After adjusting for age, sex, and season of the year, increased levels of T-tau (effect size [95% CI] of 0.343 [0.139 to 0.547]; p = 0.001) and NF-L (0.444 [0.212 to 0.676]; p = 0.001) were associated with a higher amplitude of the rest-activity rhythm. Increased fragmentation of the rhythm at baseline was associated with greater cognitive decline after one year of follow-up independent of age, sex, T-tau/Aβ42 ratio, educational level, and season of the year (- 0.715 [- 1.272 to - 0.157]; p = 0.013). Similar findings were obtained considering only the Aβ42+ patients. CONCLUSIONS Our results suggest a potential role of the circadian rest-activity pattern in predicting the cognitive decline of patients with mild-moderate AD. Further studies are warranted to confirm these findings and to elucidate whether there is causality among the observed associations.
Collapse
Affiliation(s)
- Adriano D S Targa
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, IRBLleida, Lleida, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Iván D Benítez
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, IRBLleida, Lleida, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Faridé Dakterzada
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Hospital Universitari Santa Maria, IRBLleida, Lleida, Spain
| | - John Fontenele-Araujo
- Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Olga Minguez
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, IRBLleida, Lleida, Spain
| | - Henrik Zetterberg
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ferran Barbé
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, IRBLleida, Lleida, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Hospital Universitari Santa Maria, IRBLleida, Lleida, Spain.
| |
Collapse
|
124
|
Xiao Q, Sampson JN, LaCroix AZ, Shadyab AH, Zeitzer JM, Ancoli-Israel S, Yaffe K, Stone K. Nonparametric parameters of 24-hour rest-activity rhythms and long-term cognitive decline and incident cognitive impairment in older men. J Gerontol A Biol Sci Med Sci 2021; 77:250-258. [PMID: 34558603 DOI: 10.1093/gerona/glab275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Indexed: 11/14/2022] Open
Abstract
Altered 24-hour rest-activity rhythms may be associated with cognitive impairment in older adults, but evidence from prospective studies is limited. Non-parametric methods were used to assess actigraphy-based activity patterns in 2,496 older men. Incident cognitive impairment was assessed four times over 12 years using the Modified Mini Mental State Examination (3MS) and Trails B tests, self-reported medication use, and clinical diagnosis. The highest quartile (vs. the lowest) of intradaily variability and the lowest quartiles (vs. the highest) of interdaily stability and relative amplitude were associated with incident cognitive impairment ((Hazard ratio (95% confidence interval): 1.82 (1.31, 2.53)), 1.36 (0.99, 1.86), and 1.85 (1.33, 2.56), respectively). A larger increase in intradaily variability over 7.5 years was associated with a greater subsequent decline in 3MS scores but not in Trails B performance. In conclusion, less stable and more variable rest-activity rhythms may represent early biomarkers of cognitive impairment in older men.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Epidemiology, Human Genetics and Environmental Health, School of Public Health, the University of Texas Health Science Center at Houston, Houston, TX
| | - Joshua N Sampson
- Biostatistics Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, MD
| | - Andrea Z LaCroix
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Jamie M Zeitzer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA.,Mental Illness Research Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto CA
| | - Sonia Ancoli-Israel
- Department of Psychiatry, Center for Circadian Biology, University of California, San Diego, La Jolla, CA
| | - Kristin Yaffe
- Department of Psychiatry, Neurology, and Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Katie Stone
- Research Institute, California Pacific Medical Center, San Francisco, CA
| | | |
Collapse
|
125
|
Duncan MJ, Veasey SC, Zee P. Editorial: Roles of Sleep Disruption and Circadian Rhythm Alterations on Neurodegeneration and Alzheimer's Disease. Front Neurosci 2021; 15:737895. [PMID: 34552466 PMCID: PMC8450346 DOI: 10.3389/fnins.2021.737895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- Marilyn J Duncan
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Sigrid C Veasey
- Chronobiology and Sleep Institute and Department Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Phyllis Zee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
126
|
The Effect of Bright Light Treatment on Rest-Activity Rhythms in People with Dementia: A 24-Week Cluster Randomized Controlled Trial. Clocks Sleep 2021; 3:449-464. [PMID: 34563054 PMCID: PMC8482074 DOI: 10.3390/clockssleep3030032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Bright light treatment is an effective way to influence circadian rhythms in healthy adults, but previous research with dementia patients has yielded mixed results. The present study presents a primary outcome of the DEM.LIGHT trial, a 24-week randomized controlled trial conducted at nursing homes in Bergen, Norway, investigating the effects of a bright light intervention. The intervention consisted of ceiling-mounted LED panels providing varying illuminance and correlated color temperature throughout the day, with a peak of 1000 lx, 6000 K between 10 a.m. and 3 p.m. Activity was recorded using actigraphs at baseline and after 8, 16, and 24 weeks. Non-parametric indicators and extended cosine models were used to investigate rest-activity rhythms, and outcomes were analyzed with multi-level regression models. Sixty-one patients with severe dementia (median MMSE = 4) were included. After 16 weeks, the acrophase was advanced from baseline in the intervention group compared to the control group (B = -1.02, 95%; CI = -2.00, -0.05). There was no significant difference between the groups on any other rest-activity measures. When comparing parametric and non-parametric indicators of rest-activity rhythms, 25 out of 35 comparisons were significantly correlated. The present results indicate that ambient bright light treatment did not improve rest-activity rhythms for people with dementia.
Collapse
|
127
|
Abstract
Alzheimer disease (AD) is the most common type of dementia characterized by the progressive cognitive and social decline. Clinical drug targets have heavily focused on the amyloid hypothesis, with amyloid beta (Aβ), and tau proteins as key pathophysiologic markers of AD. However, no effective treatment has been developed so far, which prompts researchers to focus on other aspects of AD beyond Aβ, and tau proteins. Additionally, there is a mounting epidemiologic evidence that various environmental factors influence the development of dementia and that dementia etiology is likely heterogenous. In the past decades, new risk factors or potential etiologies have been widely studied. Here, we review several novel epidemiologic and clinical research developments that focus on sleep, hypoxia, diet, gut microbiota, and hearing impairment and their links to AD published in recent years. At the frontiers of AD research, these findings and updates could be worthy of further attention.
Collapse
|
128
|
Li J, Somers VK, Lopez-Jimenez F, Di J, Covassin N. Demographic characteristics associated with circadian rest-activity rhythm patterns: a cross-sectional study. Int J Behav Nutr Phys Act 2021; 18:107. [PMID: 34407852 PMCID: PMC8371768 DOI: 10.1186/s12966-021-01174-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Rest-activity rhythm (RAR), a manifestation of circadian rhythms, has been associated with morbidity and mortality risk. However, RAR patterns in the general population and specifically the role of demographic characteristics in RAR pattern have not been comprehensively assessed. Therefore, we aimed to describe RAR patterns among non-institutionalized US adults and age, sex, and race/ethnicity variation using accelerometry data from a nationally representative population. METHODS This cross-sectional study was conducted using the US National Health and Nutrition Examination Survey (NHANES) 2011-2014. Participants aged ≥20 years who were enrolled in the physical activity monitoring examination and had at least four 24-h periods of valid wrist accelerometer data were included in the present analysis. 24-h RAR metrics were generated using both extended cosinor model (amplitude, mesor, acrophase and pseudo-F statistic) and nonparametric methods (interdaily stability [IS] and intradaily variability [IV]). Multivariable linear regression was used to assess the association between RAR and age, sex, and race/ethnicity. RESULTS Eight thousand two hundred participants (mean [SE] age, 49.1 [0.5] years) were included, of whom 52.2% were women and 67.3% Whites. Women had higher RAR amplitude and mesor, and also more robust (pseudo-F statistic), more stable (higher IS) and less fragmented (lower IV) RAR (all P trend < 0.001) than men. Compared with younger adults (20-39 years), older adults (≥ 60 years) exhibited reduced RAR amplitude and mesor, but more stable and less fragmented RAR, and also reached their peak activity earlier (advanced acrophase) (all P trend < 0.001). Relative to other racial/ethnic groups, Hispanics had the highest amplitude and mesor level, and most stable (highest IS) and least fragmented (lowest IV) RAR pattern (P trend < 0.001). Conversely, non-Hispanic blacks had the lowest peak activity level (lowest amplitude) and least stable (lowest IS) RAR pattern (all P trend < 0.001). CONCLUSIONS In the general adult population, RAR patterns vary significantly according to sex, age and race/ethnicity. These results may reflect demographic-dependent differences in intrinsic circadian rhythms and may have important implications for understanding racial, ethnic, sex and other disparities in morbidity and mortality risk.
Collapse
Affiliation(s)
- Jingen Li
- Department of Cardiovascular Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Francisco Lopez-Jimenez
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Junrui Di
- Department of Biostatistics, Johns Hopkins University, Baltimore, MA, 21205, USA
| | - Naima Covassin
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
129
|
Effects of Shift Work in a Sample of Italian Nurses: Analysis of Rest-Activity Circadian Rhythm. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168378. [PMID: 34444128 PMCID: PMC8391482 DOI: 10.3390/ijerph18168378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/26/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Shift work can lead to circadian desynchronization due to temporary misalignment between working hours and physiological and behavioral functioning, resulting in compromised health, insomnia, worsening of sleep quality, reduced ability to work during waking hours, and increased cardiovascular risk. We evaluated the effects of shift work on the rest-activity circadian rhythm (RAR) and health status of Italian orthopaedic nurses. The study population was 59 nurses: 44 worked the night shift and 15 worked the day shift. All carried out continuous 5-day actigraphic monitoring to assess RAR, including both the working and the rest period. The rhythmometric analysis showed that, during the working period, the night shift nurses had a significantly lower amplitude than the day shift nurses (p < 0.001), and the acrophase was significantly different between the two groups (p < 0.01). When we stratified the two groups by median body mass index (<25 kg/m2 normal weight and ≥25 kg/m2 overweight), during the working period, we noted a significantly lower amplitude for both the normal weight and the overweight nurses who worked the night shift (p < 0.01 and p < 0.001, normal weight and overweight respectively). The current findings suggest the need for further study of the relationship between activity levels and shift work.
Collapse
|
130
|
Blackman J, Swirski M, Clynes J, Harding S, Leng Y, Coulthard E. Pharmacological and non-pharmacological interventions to enhance sleep in mild cognitive impairment and mild Alzheimer's disease: A systematic review. J Sleep Res 2021; 30:e13229. [PMID: 33289311 PMCID: PMC8365694 DOI: 10.1111/jsr.13229] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022]
Abstract
Suboptimal sleep causes cognitive decline and probably accelerates Alzheimer's Disease (AD) progression. Several sleep interventions have been tested in established AD dementia cases. However early intervention is needed in the course of AD at Mild Cognitive Impairment (MCI) or mild dementia stages to help prevent decline and maintain good quality of life. This systematic review aims to summarize evidence on sleep interventions in MCI and mild AD dementia. Seven databases were systematically searched for interventional studies where ≥ 75% of participants met diagnostic criteria for MCI/mild AD dementia, with a control group and validated sleep outcome measures. Studies with a majority of participants diagnosed with Moderate to Severe AD were excluded. After removal of duplicates, 22,133 references were returned in two separate searches (August 2019 and September 2020). 325 full papers were reviewed with 18 retained. Included papers reported 16 separate studies, total sample (n = 1,056), mean age 73.5 years. 13 interventions were represented: Cognitive Behavioural Therapy - Insomnia (CBT-I), A Multi-Component Group Based Therapy, A Structured Limbs Exercise Programme, Aromatherapy, Phase Locked Loop Acoustic Stimulation, Transcranial Stimulation, Suvorexant, Melatonin, Donepezil, Galantamine, Rivastigmine, Tetrahydroaminoacridine and Continuous Positive Airway Pressure (CPAP). Psychotherapeutic approaches utilising adapted CBT-I and a Structured Limbs Exercise Programme each achieved statistically significant improvements in the Pittsburgh Sleep Quality Index with one study reporting co-existent improved actigraphy variables. Suvorexant significantly increased Total Sleep Time and Sleep Efficiency whilst reducing Wake After Sleep Onset time. Transcranial Stimulation enhanced cortical slow oscillations and spindle power during daytime naps. Melatonin significantly reduced sleep latency in two small studies and sleep to wakefulness transitions in a small sample. CPAP demonstrated efficacy in participants with Obstructive Sleep Apnoea. Evidence to support other interventions was limited. Whilst new evidence is emerging, there remains a paucity of evidence for sleep interventions in MCI and mild AD highlighting a pressing need for high quality experimental studies exploring alternative sleep interventions.
Collapse
Affiliation(s)
- Jonathan Blackman
- North Bristol NHS TrustBristolUK
- Bristol Medical SchoolUniversity of BristolBristolUK
| | | | | | | | - Yue Leng
- Department of PsychiatryUniversity of CaliforniaSan FranciscoCAUSA
| | | |
Collapse
|
131
|
Kim E, Nohara K, Wirianto M, Escobedo G, Lim JY, Morales R, Yoo SH, Chen Z. Effects of the Clock Modulator Nobiletin on Circadian Rhythms and Pathophysiology in Female Mice of an Alzheimer's Disease Model. Biomolecules 2021; 11:biom11071004. [PMID: 34356628 PMCID: PMC8301787 DOI: 10.3390/biom11071004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder and the most common cause of dementia. Various pathogenic mechanisms have been proposed to contribute to disease progression, and recent research provided evidence linking dysregulated circadian rhythms/sleep and energy metabolism with AD. Previously, we found that the natural compound Nobiletin (NOB) can directly activate circadian cellular oscillators to promote metabolic health in disease models and healthy aging in naturally aged mice. In the current study, using the amyloid-β AD model APP/PS1, we investigated circadian, metabolic and amyloid characteristics of female mice and the effects of NOB. Female APP/PS1 mice showed reduced sleep bout duration, and NOB treatment exhibited a trend to improve it. While glucose tolerance was unchanged, female APP/PS1 mice displayed exaggerated oxygen consumption and CO2 production, which was mitigated by NOB. Likewise, cold tolerance in APP/PS1 was impaired relative to WT, and interestingly was markedly enhanced in NOB-treated APP/PS1 mice. Although circadian behavioral rhythms were largely unchanged, real-time qPCR analysis revealed altered expression of several core clock genes by NOB in the cerebral cortex, notably Bmal1, Npas2, and Rora. Moreover, NOB was also able to activate various clock-controlled metabolic genes involved in insulin signaling and mitochondrial function, including Igf1, Glut1, Insr, Irs1, Ucp2, and Ucp4. Finally, we observed that NOB attenuated the expression of several AD related genes including App, Bace1, and ApoE, reduced APP protein levels, and strongly ameliorated Aβ pathology in the cortex. Collectively, these results reveal novel genotype differences and importantly beneficial effects of a natural clock-enhancing compound in biological rhythms and related pathophysiology, suggesting the circadian clock as a modifiable target for AD.
Collapse
Affiliation(s)
- Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Gabriel Escobedo
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
| | - Ji Ye Lim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
- Centro Integrativo de Biologia y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
- Correspondence:
| |
Collapse
|
132
|
McMahon M, Malneedi Y, Worthy DA, Schnyer DM. Rest-activity rhythms and white matter microstructure across the lifespan. Sleep 2021; 44:6017487. [PMID: 33269397 DOI: 10.1093/sleep/zsaa266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/09/2020] [Indexed: 11/14/2022] Open
Abstract
STUDY OBJECTIVES The purpose of this study was to examine how rest-activity (RA) rhythm stability may be associated with white matter microstructure across the lifespan in healthy adults free of significant cardiovascular risk. METHODS We analyzed multi-shell diffusion tensor images from 103 healthy young and older adults using tract-based spatial statistics (TBSS) to examine relationships between white matter microstructure and RA rhythm stability. RA measures were computed using both cosinor and non-parametric methods derived from 7 days of actigraphy data. Fractional anisotropy (FA) and mean diffusivity (MD) were examined in this analysis. Because prior studies have suggested that the corpus callosum (CC) is sensitive to sleep physiology and RA rhythms, we also conducted a focused region of interest analysis on the CC. RESULTS Greater rest-activity rhythm stability was associated with greater FA across both young and older adults, primarily in the CC and anterior corona radiata. This effect was not moderated by age group. While RA measures were associated with sleep metrics, RA rhythm measures uniquely accounted for the variance in white matter integrity. CONCLUSIONS This study strengthens existing evidence for a relationship between brain white matter structure and RA rhythm stability in the absence of health risk factors. While there are differences in RA stability between age groups, the relationship with brain white matter was present across both young and older adults. RA rhythms may be a useful biomarker of brain health across both periods of adult development.
Collapse
Affiliation(s)
- Megan McMahon
- Department of Psychology, University of Texas at Austin, Austin, TX
| | - Yoshita Malneedi
- Department of Psychology, University of Texas at Austin, Austin, TX
| | - Darrell A Worthy
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX
| | - David M Schnyer
- Department of Psychology, University of Texas at Austin, Austin, TX
| |
Collapse
|
133
|
Kim M, Liotta EM, Maas MB, Braun RI, Garcia-Canga B, Ganger DR, Ladner DP, Reid KJ, Zee PC. Rest-activity rhythm disturbance in liver cirrhosis and association with cognitive impairment. Sleep 2021; 44:6047598. [PMID: 33367862 DOI: 10.1093/sleep/zsaa288] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/20/2020] [Indexed: 11/12/2022] Open
Abstract
Cognitive impairment and disturbed sleep-wake rhythms are disabling complications of liver cirrhosis, yet there is limited understanding of how they are related. We tested the hypothesis that alterations of sleep, rest-activity, and light exposure patterns are associated with worse cognition in cirrhosis. A total of 54 ambulatory adult patients with cirrhosis and 41 age-/gender-matched healthy controls wore wrist actigraphy for rest-activity and light measurements and completed Patient-Reported Outcomes Measurement Information System sleep instruments for self-reported sleep quality. We used standard nonparametric descriptors to characterize rest-activity and light patterns, and wake after sleep onset and sleep efficiency to assess objective sleep quality. The NIH Toolbox cognition battery was used for objective cognitive evaluation using T-scores from a demographically adjusted population reference. Spearman's correlation and multivariable models were used to explore associations between measures of cognition, sleep, rest-activity, and light. Cognition was significantly impaired in cirrhosis patients. Sleep quality was worse in cirrhosis patients by subjective and objective measures compared with controls. Cirrhosis patients exhibited fragmented and dampened rest-activity rhythms, lower daytime and higher nighttime light exposure compared with controls. Worse working memory and processing speed was associated with lower daytime activity level, higher rest-activity fragmentation, lower day-to-day stability, and greater nocturnal light exposure. No association was found between cognition and sleep quality. Rest-activity fragmentation and abnormal light exposure patterns are common in patients with liver disease and are associated with the severity of cognitive impairment. Further research is needed to investigate the effects of timed bright light and exercise intervention on cognitive function in patients with liver disease.
Collapse
Affiliation(s)
- Minjee Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Chicago, IL
| | - Eric M Liotta
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Chicago, IL
- Division of Transplant, Department of Surgery, Northwestern Medicine, Chicago, IL
| | - Matthew B Maas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rosemary I Braun
- Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Biostatistics Division, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL
| | - Blas Garcia-Canga
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Daniel R Ganger
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Chicago, IL
- Division of Transplant, Department of Surgery, Northwestern Medicine, Chicago, IL
- Department of Medicine, Northwestern Medicine, Chicago, IL
| | - Daniela P Ladner
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Chicago, IL
- Division of Transplant, Department of Surgery, Northwestern Medicine, Chicago, IL
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Kathryn J Reid
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Phyllis C Zee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
- Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
134
|
Obayashi K, Saeki K, Yamagami Y, Kurumatani N, Sugie K, Kataoka H. Circadian activity rhythm in Parkinson's disease: findings from the PHASE study. Sleep Med 2021; 85:8-14. [PMID: 34265483 DOI: 10.1016/j.sleep.2021.06.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Circadian disruptions in Parkinson's disease (PD) are characterized as amplitude reduction rather than as phase shift; however, large-scale studies evaluating circadian rhythms between PD patients and non-PD older adults have not been performed. The present study aimed to compare the circadian activity rhythm (CAR) between PD patients and non-PD older adults. METHODS In this cross-sectional study on 157 PD outpatients and 1111 community-dwelling older adults (controls), physical activity was measured using actigraphy at 1-min intervals over 6 days in PD patients and 2 days in non-PD older adults. Data were base-10 log-transformed and regretted to the sigmoidally transformed cosine curve. RESULTS The mean amplitude (log counts/min) and acrophase were 1.85 (SD, 0.52) and 14:19 (SD, 1:15), respectively, in the controls (n = 1111); 1.42 (0.48) and 14:24 (1:20), respectively, in the early-stage (Hoehn-Yahr I and II) PD patients (n = 95); and 1.23 (0.54) and 13:41 (1:56), respectively, in the late-stage (Hoehn-Yahr III-V) PD patients (n = 62). Multivariable analysis revealed significantly lower amplitude in the early-stage and late-stage PD groups than in the controls. The acrophase significantly advanced in the late-stage PD group than in the controls. With the advancement of PD stage, amplitude and peak significantly decreased; trough increased; acrophase and active offset advanced; and robustness weakened. CONCLUSIONS Compared with non-PD older adults, PD patients exhibited a phase advance in CAR, along with amplitude reduction. With an advanced stage of PD, a phase advance in CAR also occurred, along with amplitude reduction and weakened robustness.
Collapse
Affiliation(s)
- Kenji Obayashi
- Department of Epidemiology, Nara Medical University School of Medicine, Nara, Japan.
| | - Keigo Saeki
- Department of Epidemiology, Nara Medical University School of Medicine, Nara, Japan
| | - Yuki Yamagami
- Department of Epidemiology, Nara Medical University School of Medicine, Nara, Japan
| | - Norio Kurumatani
- Department of Epidemiology, Nara Medical University School of Medicine, Nara, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University School of Medicine, Nara, Japan
| | - Hiroshi Kataoka
- Department of Neurology, Nara Medical University School of Medicine, Nara, Japan.
| |
Collapse
|
135
|
Leng Y, Blackwell T, Cawthon PM, Ancoli-Israel S, Stone KL, Yaffe K. Association of Circadian Abnormalities in Older Adults With an Increased Risk of Developing Parkinson Disease. JAMA Neurol 2021; 77:1270-1278. [PMID: 32539075 DOI: 10.1001/jamaneurol.2020.1623] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Importance Disruption in circadian activity rhythms is very common in older adults, particularly among those with neurodegenerative diseases, including Parkinson disease (PD). However, whether circadian disruption could be a prodrome for PD is unclear. Objective To determine the association between rest-activity rhythm (RAR) and risk of incident PD and to explore whether this association is independent of nighttime sleep disturbances. Design, Setting, and Participants The ancillary sleep study of the longitudinal cohort Osteoporotic Fractures in Men Study (MrOS) was conducted from December 1, 2003, to March 31, 2005. Of the 3135 community-dwelling men enrolled in the MrOS sleep study, 3049 had technically adequate RAR data; of these, 119 were excluded for having prevalent PD or missing incident data, leaving 2930 men without PD at baseline. Data were analyzed from February 1 through August 31, 2019. Exposures Twenty four-hour RAR parameters (amplitude, mesor, robustness, and acrophase) generated by wrist actigraphy-extended cosinor analysis. Main Outcomes and Measures Incident PD based on physician diagnosis. Multivariable logistic regression was used to determine the association between quartiles of RAR parameters and risk of incident PD. Results Among the 2930 men included in the analysis (mean [SD] age, 76.3 [5.5] years), 78 (2.7%) developed PD during 11 years of follow-up. After accounting for all covariates, the risk of PD increased with decreasing circadian amplitude (strength of the rhythm) (odds ratio [OR] per 1-SD decrease, 1.77; 95% CI, 1.30-2.41), mesor (mean level of activity) (OR per 1-SD decrease, 1.64; 95% CI, 1.22-2.21), or robustness (how closely activity follows a cosine 24-hour pattern) (OR per 1-SD decrease, 1.54; 95% CI, 1.14-2.07) (P < .005 for trend). Those in the lowest quartile of amplitude, mesor, or robustness had approximately 3 times the risk of developing PD compared with those in the highest quartile of amplitude (OR, 3.11; 95% CI, 1.54-6.29), mesor (OR, 3.04; 95% CI, 1.54-6.01), and robustness (OR, 2.65; 95% CI, 1.24-5.66). The association remained after further adjustment for nighttime sleep disturbances and duration in the lowest compared with the highest quartile (OR for amplitude, 3.56 [95% CI, 1.68-7.56]; OR for mesor, 3.24 [95% CI, 1.52-6.92]; and OR for robustness, 3.34 [95% CI, 1.45-7.67]). These associations were somewhat attenuated, but the pattern remained similar after excluding PD cases developed within 2 years after baseline in the lowest compared with the highest quartile (OR for amplitude, 2.40 [95% CI, 1.15-5.00]; OR for mesor, 2.76 [95% CI, 1.35-5.67]; and OR for robustness, 2.33 [95% CI, 1.07-5.07]). Acrophase was not significantly associated with risk of PD. Conclusions and Relevance In this cohort study, reduced circadian rhythmicity was associated with an increased risk of incident PD, suggesting it may represent an important prodromal feature for PD. Future studies are needed to determine whether circadian disruption could also be a risk factor for PD and whether strategies to improve circadian function affect the risk of PD.
Collapse
Affiliation(s)
- Yue Leng
- Department of Psychiatry, University of California, San Francisco
| | - Terri Blackwell
- California Pacific Medical Center Research Institute, California Pacific Medical Center, San Francisco
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, California Pacific Medical Center, San Francisco
| | | | - Katie L Stone
- California Pacific Medical Center Research Institute, California Pacific Medical Center, San Francisco
| | - Kristine Yaffe
- Department of Psychiatry, San Francisco Veterans Affairs Medical Center, University of California, San Francisco.,Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco.,Department of Epidemiology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco
| |
Collapse
|
136
|
Impact of Shift Work and Long Working Hours on Worker Cognitive Functions: Current Evidence and Future Research Needs. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18126540. [PMID: 34204504 PMCID: PMC8296479 DOI: 10.3390/ijerph18126540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022]
Abstract
Particular working conditions and/or organization of working time may cause important sleep disturbances that have been proposed to be predictive of cognitive decline. In this regard, circadian rhythm misalignment induced by exposure to night work or long working hours would be responsible for cognitive impairment. Nevertheless, evidence supporting this correlation is limited and several issues still need to be elucidated. In this regard, we conducted a systematic review to evaluate the association between shift/night work and cognitive impairment and address its main determinants. Information provided by the reviewed studies suggested that night work might have serious immediate negative effects especially on cognitive domains related to attention, memory and response inhibition. Furthermore, cognitive performance would progressively worsen over consecutive night shifts or following exposure to very long work shifts. Otherwise, conflicting results emerged regarding the possible etiological role that night work chronic exposure would have on cognitive impairment. Therefore, circadian rhythm desynchronization, lack of sleep and fatigue resulting from night work may negatively impact worker’s cognitive efficiency. However, in light of the considerable methodological variability of the reviewed studies, we proposed to develop a standardized research and evaluation strategy in order to obtain a better and comprehensive understanding of this topic.
Collapse
|
137
|
Bang E, Tobery A, Montgomery KS, Fincher AS, Earnest DJ, Murchison DA, Griffith WH. Amitriptyline Decreases GABAergic Transmission in Basal Forebrain Neurons Using an Optogenetic Model of Aging. Front Aging Neurosci 2021; 13:673155. [PMID: 34122049 PMCID: PMC8193944 DOI: 10.3389/fnagi.2021.673155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
The antidepressant drug amitriptyline is used in the treatment of clinical depression and a variety of neurological conditions such as anxiety, neuropathic pain disorders and migraine. Antidepressants are associated with both therapeutic and untoward effects, and their use in the elderly has tripled since the mid-1990s. Because of this widespread use, we are interested in testing the acute effects of amitriptyline on synaptic transmission at therapeutic concentrations well below those that block voltage-gated calcium channels. We found that 3 μM amitriptyline reduced the frequency of spontaneous GABAergic inhibitory postsynaptic currents (IPSCs) and reduced quantal content in mice at ages of 7-10 mo. and 23-25 mo., suggesting a presynaptic mechanism of action that does not diminish with age. We employed a reduced synaptic preparation of the basal forebrain (BF) and a new optogenetic aging model utilizing a bacterial artificial chromosome (BAC) transgenic mouse line with stable expression of the channelrhodopsin-2 (ChR2) variant H134R specific for GABAergic neurons [VGAT-ChR2(H134R)-EYFP]. This model enables optogenetic light stimulation of specific GABAergic synaptic terminals across aging. Age-related impairment of circadian behavior was used to confirm predictable age-related changes associated with this model. Our results suggest that low concentrations of amitriptyline act presynaptically to reduce neurotransmitter release and that this action is maintained during aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - William H. Griffith
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
138
|
Duffy JF, Abbott SM, Burgess HJ, Crowley SJ, Emens JS, Epstein LJ, Gamble KL, Hasler BP, Kristo DA, Malkani RG, Rahman SA, Thomas SJ, Wyatt JK, Zee PC, Klerman EB. Workshop report. Circadian rhythm sleep-wake disorders: gaps and opportunities. Sleep 2021; 44:zsaa281. [PMID: 33582815 PMCID: PMC8120340 DOI: 10.1093/sleep/zsaa281] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/02/2020] [Indexed: 01/09/2023] Open
Abstract
This White Paper presents the results from a workshop cosponsored by the Sleep Research Society (SRS) and the Society for Research on Biological Rhythms (SRBR) whose goals were to bring together sleep clinicians and sleep and circadian rhythm researchers to identify existing gaps in diagnosis and treatment and areas of high-priority research in circadian rhythm sleep-wake disorders (CRSWD). CRSWD are a distinct class of sleep disorders caused by alterations of the circadian time-keeping system, its entrainment mechanisms, or a misalignment of the endogenous circadian rhythm and the external environment. In these disorders, the timing of the primary sleep episode is either earlier or later than desired, irregular from day-to-day, and/or sleep occurs at the wrong circadian time. While there are incomplete and insufficient prevalence data, CRSWD likely affect at least 800,000 and perhaps as many as 3 million individuals in the United States, and if Shift Work Disorder and Jet Lag are included, then many millions more are impacted. The SRS Advocacy Taskforce has identified CRSWD as a class of sleep disorders for which additional high-quality research could have a significant impact to improve patient care. Participants were selected for their expertise and were assigned to one of three working groups: Phase Disorders, Entrainment Disorders, and Other. Each working group presented a summary of the current state of the science for their specific CRSWD area, followed by discussion from all participants. The outcome of those presentations and discussions are presented here.
Collapse
Affiliation(s)
- Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Sabra M Abbott
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Helen J Burgess
- Department of Psychiatry, University of Michigan, Ann Arbor, MI
| | - Stephanie J Crowley
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL
| | - Jonathan S Emens
- Department of Psychiatry, Oregon Health & Science University, Portland, OR
| | - Lawrence J Epstein
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Karen L Gamble
- Department of Psychiatry University of Alabama at Birmingham, Birmingham, AL
| | - Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - David A Kristo
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Roneil G Malkani
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shadab A Rahman
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - S Justin Thomas
- Department of Psychiatry University of Alabama at Birmingham, Birmingham, AL
| | - James K Wyatt
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL
| | - Phyllis C Zee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
139
|
Falck RS, Davis JC, Best JR, Chan PCY, Li LC, Wyrough AB, Bennett KJ, Backhouse D, Liu-Ambrose T. Effect of a Multimodal Lifestyle Intervention on Sleep and Cognitive Function in Older Adults with Probable Mild Cognitive Impairment and Poor Sleep: A Randomized Clinical Trial. J Alzheimers Dis 2021; 76:179-193. [PMID: 32444553 DOI: 10.3233/jad-200383] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Poor sleep is common among older adults with mild cognitive impairment (MCI) and may contribute to further cognitive decline. Whether multimodal lifestyle intervention that combines bright light therapy (BLT), physical activity (PA), and good sleep hygiene can improve sleep in older adults with MCI and poor sleep is unknown. OBJECTIVE To assess the effect of a multimodal lifestyle intervention on sleep in older adults with probable MCI and poor sleep. METHODS This was a 24-week proof-of-concept randomized trial of 96 community-dwelling older adults aged 65-85 years with probable MCI (<26/30 on the Montreal Cognitive Assessment) and poor sleep (>5 on the Pittsburgh Sleep Quality Index [PSQI]). Participants were allocated to either a multimodal lifestyle intervention (INT); or 2) education + attentional control (CON). INT participants received four once-weekly general sleep hygiene education classes, followed by 20-weeks of: 1) individually-timed BLT; and 2) individually-tailored PA promotion. Our primary outcome was sleep efficiency measured using the MotionWatch8© (MW8). Secondary outcomes were MW8-measured sleep duration, fragmentation index, wake-after-sleep-onset, latency, and PSQI-measured subjective sleep quality. RESULTS There were no significant between-group differences in MW8 measured sleep efficiency at 24-weeks (estimated mean difference [INT -CON]: 1.18%; 95% CI [-0.99, 3.34]), or any other objective-estimate of sleep. However, INT participants reported significantly better subjective sleep quality at 24-weeks (estimated mean difference: -1.39; 95% CI [-2.72, -0.06]) compared to CON. CONCLUSION Among individuals with probable MCI and poor sleep, a multimodal lifestyle intervention improves subjective sleep quality, but not objectively estimated sleep.
Collapse
Affiliation(s)
- Ryan S Falck
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer C Davis
- Faculty of Management, University of British Columbia-Okanagan Campus, Kelowna, British Columbia, Canada
| | - John R Best
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Patrick C Y Chan
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Linda C Li
- Arthritis Research Canada, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anne B Wyrough
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kimberly J Bennett
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Backhouse
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Teresa Liu-Ambrose
- Aging, Mobility, and Cognitive Neuroscience Lab, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute. University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
140
|
Association between sleep quality and subjective cognitive decline: evidence from a community health survey. Sleep Med 2021; 83:123-131. [PMID: 33993029 DOI: 10.1016/j.sleep.2021.04.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Little is known concerning whether subjective cognitive decline (SCD) is associated with sleep quality. This study aimed to identify the association between self-reported quality of sleep and SCD in a large population of middle-aged and older adults in Korea. METHODS We conducted this study based on data collected from the 2018 Korean Community Health Survey. Individuals aged 40 years and older who responded to the Behavioral Risk Factor Surveillance System (BRFSS) and Pittsburgh Sleep Quality Index (PSQI) assessments and did not lack data about multiple covariates were included. A total of 37,712 respondents with SCD and 135,119 those without SCD were included. Sleep quality was estimated using the PSQI, which includes seven self-reported components for sleep health assessment. SCD was assessed using the BRFSS. Logistic regression models adjusted for confounders were used to examine whether each component of the sleep quality index was related to SCD. Additional analysis of the correlation between quantified scores for each component and SCD-related functional limitations as ordinal variables was performed. RESULTS The mean age was 62.7 years in the SCD group and 56.4 years in the control group. In total, 13,777 (28.9%) respondents were male in the SCD group and 62,439 (50.7%) in the control group. The adjusted odds ratios of SCD were 1.25 for very bad sleep quality, 1.26 for long sleep latency, 1.16 for <5 h of sleep duration, 1.08 for <65% habitual sleep efficiency, 2.29 for high sleep disturbance, 1.26 for use of sleep medication ≥3 times a week, and 2.47 for high daytime dysfunction due to sleep problems compared to good sleep conditions. Furthermore, a higher score for each component of the sleep quality index correlated with greater SCD-related functional limitations. CONCLUSIONS Our study provides evidence that poor sleep quality is closely related to both SCD and SCD-related functional limitations.
Collapse
|
141
|
Alfini A, Albert M, Faria AV, Soldan A, Pettigrew C, Wanigatunga S, Zipunnikov V, Spira AP. Associations of actigraphic sleep and circadian rest/activity rhythms with cognition in the early phase of Alzheimer's disease. ACTA ACUST UNITED AC 2021; 2:zpab007. [PMID: 34095836 PMCID: PMC8168567 DOI: 10.1093/sleepadvances/zpab007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Indexed: 11/20/2022]
Abstract
Study Objectives To compare sleep and circadian rest/activity rhythms (RARs), quantified by standard and novel actigraphic metrics, between controls and participants with mild cognitive impairment (MCI), and to examine the cross-sectional relationships between these measures and cognition. Methods Actigraphy data were collected in 179 older individuals (mean age = 72.6 years) with normal cognition (n = 153) and MCI (n = 26). Sleep parameters (e.g. sleep efficiency), and standard nonparametric RARs (e.g. interdaily stability) were generated. Functional principal component analysis (fPCA) was used to generate three novel RAR metrics (fPC1, fPC2, and fPC3). Cognitive composite scores reflecting episodic memory and executive function were derived using factor analysis. Regression models compared sleep and RAR parameters between diagnostic groups and their association with cognitive performance. Results Compared to controls, the MCI group exhibited lower levels of the standard RAR parameter: relative amplitude and fPC3—a novel RAR whereby lower scores reflected a lower rhythm peak, as well as greater nighttime activity and less activity in the morning. Across groups, several standard RAR parameters (e.g. interdaily stability) and fPC3 were associated with better episodic memory and executive function performance. Additionally, several standard RAR measures (e.g. relative amplitude) and the novel RAR measure fPC1 (reflecting the total volume of activity and rhythm strength) were associated with better executive function performance. Conclusions Individuals with MCI have altered circadian RARs compared to controls, including the novel RAR metric fPC3, reflecting greater nighttime activity and less activity in the morning compared to mean values. Additionally, these measures are significantly associated with cognitive performance.
Collapse
Affiliation(s)
- Alfonso Alfini
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Andreia V Faria
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD
| | - Anja Soldan
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sarah Wanigatunga
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Vadim Zipunnikov
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
142
|
Wallace ML, Yu L, Buysse DJ, Stone KL, Redline S, Smagula SF, Stefanick ML, Kritz-Silverstein D, Hall MH. Multidimensional sleep health domains in older men and women: an actigraphy factor analysis. Sleep 2021; 44:5904464. [PMID: 32918075 DOI: 10.1093/sleep/zsaa181] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/11/2020] [Indexed: 12/31/2022] Open
Abstract
The multidimensional sleep health framework emphasizes that sleep can be characterized across several domains, with implications for developing novel sleep treatments and improved prediction and health screening. However, empirical evidence regarding the domains and representative measures that exist in actigraphy-assessed sleep is lacking. We aimed to establish these domains and representative measures in older adults by examining the factor structure of 28 actigraphy-derived sleep measures from 2,841 older men from the Osteoporotic Fractures in Men Sleep Study and, separately, from 2,719 older women from the Study of Osteoporotic Fractures. Measures included means and standard deviations of actigraphy summary measures and estimates from extended cosine models of the raw actigraphy data. Exploratory factor analyses revealed the same five factors in both sexes: Timing (e.g. mean midpoint from sleep onset to wake-up), Efficiency (e.g. mean sleep efficiency), Duration (e.g. mean minutes from sleep onset to wake-up), Sleepiness/Wakefulness (e.g. mean minutes napping and amplitude of rhythm), and Regularity (e.g. standard deviation of the midpoint). Within each sex, confirmatory factor analyses confirmed the one-factor structure of each factor and the entire five-factor structure (Comparative Fit Index and Tucker-Lewis Index ≥ 0.95; Root Mean Square Error of Approximation 0.08-0.38). Correlation magnitudes among factors ranged from 0.01 to 0.34. These findings demonstrate the validity of conceptualizing actigraphy sleep as multidimensional, provide a framework for selecting sleep health domains and representative measures, and suggest targets for behavioral interventions. Similar analyses should be performed with additional measures of rhythmicity, other age ranges, and more racially/ethnically diverse samples.
Collapse
Affiliation(s)
| | - Lan Yu
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Daniel J Buysse
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Susan Redline
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - Marcia L Stefanick
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA
| | - Donna Kritz-Silverstein
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA
| | - Martica H Hall
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
143
|
Kim REY, Kim HJ, Kim S, Abbott RD, Thomas RJ, Yun CH, Lee HW, Shin C. A longitudinal observational population-based study of brain volume associated with changes in sleep timing from middle to late-life. Sleep 2021; 44:5973752. [PMID: 33170277 DOI: 10.1093/sleep/zsaa233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 10/06/2020] [Indexed: 01/22/2023] Open
Abstract
STUDY OBJECTIVES Sleep behaviors are related to brain structure and function, but the impact of long-term changes in sleep timing on brain health has not been clearly addressed. The purpose of this study was to examine the association of longitudinal changes in sleep timing from middle to late-life with gray matter volume (GMV), an important marker of brain aging. METHODS We enrolled 1798 adults (aged 49-82 years, men 54.6%) who underwent magnetic resonance imaging (MRI) between 2011 and 2014. Midsleep time (MST) on free days corrected for sleep debt on workdays was adopted as a marker of sleep timing. Data on MST were available at the time of MRI assessment and at examinations that were given 9 years earlier (2003-2004). Longitudinal changes in MST over the 9-year period were derived and categorized into quartiles. Subjects in quartile 1 were defined as "advancers" (MST advanced ≥ 1 h) while those in quartile 4 were defined as "delayers" (MST delayed ≥ 0.2 h). Quartiles 2-3 defined a reference group (MST change was considered modest). The relationship of GMV with MST changes over 9 years was investigated. RESULTS Nine-year change in MST were significantly associated with GMV. Compared to the reference group, advancers had smaller GMVs in the frontal and temporal regions. A delay in MST was also associated with smaller cerebellar GMV. CONCLUSIONS In middle-to-late adulthood, the direction of change in MST is associated with GMV. While advancers and delayers in MST tend to present lower GMV, associations appear to differ across brain regions.
Collapse
Affiliation(s)
- Regina E Y Kim
- College of Medicine, Korea University, Republic of Korea.,College of Psychiatry, University of Iowa, Iowa City, IA
| | - Hyeon Jin Kim
- Department of Neurology and Medical Science, School of Medicine, Ewha Woman University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea
| | - Soriul Kim
- College of Medicine, Korea University, Republic of Korea
| | | | - Robert J Thomas
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center and the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA
| | - Chang-Ho Yun
- Department of Neurology, Seoul National University Bundang Hospital, Republic of Korea
| | - Hyang Woon Lee
- Department of Neurology and Medical Science, School of Medicine, Ewha Woman University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea.,Department of Computational Medicine, System Health & Engineering Major in Graduate School (BK21 Plus Program), Ewha Womans University, Seoul, Republic of Korea
| | - Chol Shin
- College of Medicine, Korea University, Republic of Korea
| |
Collapse
|
144
|
Cordone S, Scarpelli S, Alfonsi V, De Gennaro L, Gorgoni M. Sleep-Based Interventions in Alzheimer's Disease: Promising Approaches from Prevention to Treatment along the Disease Trajectory. Pharmaceuticals (Basel) 2021; 14:383. [PMID: 33921870 PMCID: PMC8073746 DOI: 10.3390/ph14040383] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
The multifactorial nature of Alzheimer's disease (AD) has led scientific researchers to focus on the modifiable and treatable risk factors of AD. Sleep fits into this context, given the bidirectional relationship with AD confirmed by several studies over the last years. Sleep disorders appear at an early stage of AD and continue throughout the entire course of the pathology. Specifically, sleep abnormalities, such as more fragmented sleep, increase in time of awakenings, worsening of sleep quality and primary sleep disorders raise with the severity and progression of AD. Intervening on sleep, therefore, means acting both with prevention strategies in the pre-clinical phase and with treatments during the course of the disease. This review explores sleep disturbances in the different stages of AD, starting from the pre-clinical stage. Particular attention is given to the empirical evidence investigating obstructive sleep apnea (OSA) disorder and the mechanisms overlapping and sharing with AD. Next, we discuss sleep-based intervention strategies in the healthy elderly population, mild cognitive impairment (MCI) and AD patients. We mention interventions related to behavioral strategies, combination therapies, and bright light therapy, leaving extensive space for new and raising evidence on continuous positive air pressure (CPAP) treatment effectiveness. Finally, we clarify the role of NREM sleep across the AD trajectory and consider the most recent studies based on the promising results of NREM sleep enhancement, which use innovative experimental designs and techniques.
Collapse
Affiliation(s)
- Susanna Cordone
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Serena Scarpelli
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| | | | - Luigi De Gennaro
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Maurizio Gorgoni
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| |
Collapse
|
145
|
Deciphering the Interacting Mechanisms of Circadian Disruption and Alzheimer's Disease. Neurochem Res 2021; 46:1603-1617. [PMID: 33871799 DOI: 10.1007/s11064-021-03325-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 03/21/2021] [Accepted: 04/09/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is one of the crucial causative factors for progressive dementia. Neuropathologically, AD is characterized by the extracellular accumulation of amyloid beta plaques and intracellular neurofibrillary tangles in cortical and limbic regions of the human brain. The circadian system is one of the many affected physiological processes in AD, the dysfunction of which may reflect in the irregularity of the sleep/wake cycle. The interplay of circadian and sleep disturbances inducing AD progression is bidirectional. Sleep-associated pathological alterations are frequently evident in AD. Understanding the interrelation between circadian disruption and AD may allow for earlier identification of AD pathogenesis as well as better suited approaches and potential therapies to combat dementia. In this article, we examine the existing literature related to the molecular mechanisms of the circadian clock and interacting mechanisms of circadian disruption and AD pathogenesis.
Collapse
|
146
|
Yi Lee PM, Ling Kwok BH, Ting Ma JY, Tse LA. A population-based prospective study on rest-activity rhythm and mild cognitive impairment among Hong Kong healthy community-dwelling older adults. Neurobiol Sleep Circadian Rhythms 2021; 10:100065. [PMID: 33997474 PMCID: PMC8091051 DOI: 10.1016/j.nbscr.2021.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 02/28/2021] [Accepted: 04/06/2021] [Indexed: 01/13/2023] Open
Abstract
Background Relatively few studies investigated the association between rest-activity circadian rhythm and cognitive impairment in population-based study, and the evidence from Asian populations is sparse. We aimed to examine the relationship of actigraphy measured rest-activity circadian rhythm with mild cognitive impairment (MCI) or cognitive impairment in Hong Kong healthy community-dwelling older adults. Methods We recruited 174 Hong Kong healthy adults aged ≥65 years (36 male vs. 138 female) during April-September 2018, and followed up them for 12 months. Participants were invited to wear wrist actigraphy for 7 days in both baseline and follow-up study. We used the actigraph data to calculate their midline statistic of rhythm (MESOR), amplitude, acrophase and percent rhythm. Montreal Cognitive Assessment (MoCA) was used to assess their cognitive scores at baseline and follow-up. Multivariate logistic regression model was performed to estimate the association of rest-activity circadian rhythm parameters with MCI; whilst multinomial logistic regression model was used to examine the association between rhythm parameters and changes of cognitive scores (i.e., worsen: <-1, stable: -1 to 1, better cognition: ≥2) after 12-months follow-up respectively. Results There was no association between rest-activity circadian rhythm parameters and MCI or cognitive impairment at baseline. Compared to those with an averaged value of acrophase (1:24pm-3:00pm), results of multinominal logistic regression showed that participants with a delayed acrophase (after 3:00pm) were less likely to have better cognition (adjusted odds ratio (AOR) = 0.32, 95% confidence interval (CI) = 0.11-0.88). Upon one year of follow-up, participants who delayed their acrophase for 24 min than their baseline measurements were also less likely to have better cognitive functions (AOR = 0.26, 95%CI = 0.08-0.79). Conclusions Results from both the baseline survey and follow-up study consistently confirmed that older adults, especially in light of the majority of participants being the females, with delayed acrophase were less likely to have better cognition in the Asian population.
Collapse
Affiliation(s)
- Priscilla Ming Yi Lee
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Bonnie Ho Ling Kwok
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Julie Yuen Ting Ma
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Lap Ah Tse
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
147
|
Ruby NF. Suppression of Circadian Timing and Its Impact on the Hippocampus. Front Neurosci 2021; 15:642376. [PMID: 33897354 PMCID: PMC8060574 DOI: 10.3389/fnins.2021.642376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/17/2021] [Indexed: 01/02/2023] Open
Abstract
In this article, I describe the development of the disruptive phase shift (DPS) protocol and its utility for studying how circadian dysfunction impacts memory processing in the hippocampus. The suprachiasmatic nucleus (SCN) of the Siberian hamster is a labile circadian pacemaker that is easily rendered arrhythmic (ARR) by a simple manipulation of ambient lighting. The DPS protocol uses room lighting to administer a phase-advancing signal followed by a phase-delaying signal within one circadian cycle to suppress clock gene rhythms in the SCN. The main advantage of this model for inducing arrhythmia is that the DPS protocol is non-invasive; circadian rhythms are eliminated while leaving the animals neurologically and genetically intact. In the area of learning and memory, DPS arrhythmia produces much different results than arrhythmia by surgical ablation of the SCN. As I show, SCN ablation has little to no effect on memory. By contrast, DPS hamsters have an intact, but arrhythmic, SCN which produces severe deficits in memory tasks that are accompanied by fragmentation of electroencephalographic theta oscillations, increased synaptic inhibition in hippocampal circuits, and diminished responsiveness to cholinergic signaling in the dentate gyrus of the hippocampus. The studies reviewed here show that DPS hamsters are a promising model for translational studies of adult onset circadian dysfunction in humans.
Collapse
Affiliation(s)
- Norman F. Ruby
- Biology Department, Stanford University, Stanford, CA, United States
| |
Collapse
|
148
|
Tabuchi M, Coates KE, Bautista OB, Zukowski LH. Light/Clock Influences Membrane Potential Dynamics to Regulate Sleep States. Front Neurol 2021; 12:625369. [PMID: 33854471 PMCID: PMC8039321 DOI: 10.3389/fneur.2021.625369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
The circadian rhythm is a fundamental process that regulates the sleep-wake cycle. This rhythm is regulated by core clock genes that oscillate to create a physiological rhythm of circadian neuronal activity. However, we do not know much about the mechanism by which circadian inputs influence neurons involved in sleep-wake architecture. One possible mechanism involves the photoreceptor cryptochrome (CRY). In Drosophila, CRY is receptive to blue light and resets the circadian rhythm. CRY also influences membrane potential dynamics that regulate neural activity of circadian clock neurons in Drosophila, including the temporal structure in sequences of spikes, by interacting with subunits of the voltage-dependent potassium channel. Moreover, several core clock molecules interact with voltage-dependent/independent channels, channel-binding protein, and subunits of the electrogenic ion pump. These components cooperatively regulate mechanisms that translate circadian photoreception and the timing of clock genes into changes in membrane excitability, such as neural firing activity and polarization sensitivity. In clock neurons expressing CRY, these mechanisms also influence synaptic plasticity. In this review, we propose that membrane potential dynamics created by circadian photoreception and core clock molecules are critical for generating the set point of synaptic plasticity that depend on neural coding. In this way, membrane potential dynamics drive formation of baseline sleep architecture, light-driven arousal, and memory processing. We also discuss the machinery that coordinates membrane excitability in circadian networks found in Drosophila, and we compare this machinery to that found in mammalian systems. Based on this body of work, we propose future studies that can better delineate how neural codes impact molecular/cellular signaling and contribute to sleep, memory processing, and neurological disorders.
Collapse
Affiliation(s)
- Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | | | | |
Collapse
|
149
|
Effects of long sleep time and irregular sleep-wake rhythm on cognitive function in older people. Sci Rep 2021; 11:7039. [PMID: 33782431 PMCID: PMC8007758 DOI: 10.1038/s41598-021-85817-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/24/2021] [Indexed: 01/13/2023] Open
Abstract
Sleep disturbances and cognitive decline are common in older adults. We aimed to investigate the effects of the total sleep time (TST) and sleep–wake rhythm on executive function and working memory in older adults. In 63 older participants, we measured the TST, wake after sleep onset (WASO), and sleep timing (midpoint between bedtime and wake-up time) using actigraphy. Executive function was evaluated with the trail making test B (TMT-B) and Wisconsin card sorting test (WCST). The number of back task (N-back task) was used to measure working memory. Participants with a TST ≥ 8 h had a significantly lower percentage of correct answers (% correct) on the 1-back task than those with a TST < 8 h. The % correct on the 1-back task was significantly correlated with the TST, WASO, and sleep timing. Multiple regression analyses revealed that the TST and sleep timing were significant factors of the % correct on the 1-back task. The TMT-B score was significantly correlated with the sleep timing. Category achievement on the WCST was significantly correlated with the standard deviation of the sleep timing. Therefore, a long sleep time and an irregular sleep–wake rhythm could have adverse effects on executive function and working memory in older people.
Collapse
|
150
|
Montaruli A, Castelli L, Mulè A, Scurati R, Esposito F, Galasso L, Roveda E. Biological Rhythm and Chronotype: New Perspectives in Health. Biomolecules 2021; 11:biom11040487. [PMID: 33804974 PMCID: PMC8063933 DOI: 10.3390/biom11040487] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
The circadian rhythm plays a fundamental role in regulating biological functions, including sleep–wake preference, body temperature, hormonal secretion, food intake, and cognitive and physical performance. Alterations in circadian rhythm can lead to chronic disease and impaired sleep. The circadian rhythmicity in human beings is represented by a complex phenotype. Indeed, over a 24-h period, a person’s preferred time to be more active or to sleep can be expressed in the concept of morningness–eveningness. Three chronotypes are distinguished: Morning, Neither, and Evening-types. Interindividual differences in chronotypes need to be considered to reduce the negative effects of circadian disruptions on health. In the present review, we examine the bi-directional influences of the rest–activity circadian rhythm and sleep–wake cycle in chronic pathologies and disorders. We analyze the concept and the main characteristics of the three chronotypes.
Collapse
Affiliation(s)
- Angela Montaruli
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
- IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| | - Lucia Castelli
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
| | - Antonino Mulè
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
| | - Raffaele Scurati
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
| | - Fabio Esposito
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
- IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| | - Letizia Galasso
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
- Correspondence: ; Tel.: +2-5031-4656
| | - Eliana Roveda
- Department of Biomedical Sciences for Health, University of Milan, Via G. Colombo 71, 20133 Milan, Italy; (A.M.); (L.C.); (A.M.); (R.S.); (F.E.); (E.R.)
- IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| |
Collapse
|