1
|
Ma C, Shen B, Chen L, Yang G. Impacts of circadian disruptions on behavioral rhythms in mice. FASEB J 2024; 38:e70183. [PMID: 39570004 DOI: 10.1096/fj.202401536r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Circadian rhythms are fundamental biological processes that recur approximately every 24 h, with the sleep-wake cycle or circadian behavior being a well-known example. In the field of chronobiology, mice serve as valuable model animals for studying mammalian circadian rhythms due to their genetic similarity to humans and the availability of various genetic tools for manipulation. Monitoring locomotor activity in mice provides valuable insights into the impact of various conditions or disturbances on circadian behavior. In this review, we summarized the effects of disturbance of biological rhythms on circadian behavior in mice. External factors, especially light exert a significant impact on circadian behavior. Additionally, feeding timing, food composition, ambient temperature, and physical exercise contribute to variations in the behavior of the mouse. Internal factors, including gender, age, genetic background, and clock gene mutation or deletion, are effective as well. Understanding the effects of circadian disturbances on murine behavior is essential for gaining insights into the underlying mechanisms of circadian regulation and developing potential therapeutic interventions for circadian-related disorders in humans.
Collapse
Affiliation(s)
- Changxiao Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
2
|
Chen D, Qian S, Qian W, Wu M, Wang X, Shen H, Long X, Ye M, Gong Y, Chen G. Repetitive Transcranial Magnetic Stimulation Alleviates MPTP-Induced Parkinson's Disease Symptoms by Regulating CaMKII-CREB-BMAL1 Pathway in Mice Model. Neuropsychiatr Dis Treat 2024; 20:1693-1710. [PMID: 39279880 PMCID: PMC11402372 DOI: 10.2147/ndt.s465898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024] Open
Abstract
Background Repetitive transcranial magnetic stimulation (rTMS) is a noninvasive neuromodulation technique that shows promise for the treatment of Parkinson's disease (PD). However, there is still limited understanding of the optimal stimulation frequencies and whether rTMS can alleviate PD symptoms by regulating the CaMKII-CREB-BMAL1 pathway. Methods A PD mouse model was induced intraperitoneally with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and treated with 1 Hz, 5 Hz, and 10 Hz rTMS. The neurological function, survival of dopaminergic neurons, and protein levels of Tyrosine hydroxylase (TH), α-synuclein(α-syn), and brain-derived neurotrophic factor (BDNF) in the striatum were measured to determine the optimal stimulation frequencies of rTMS treatment in PD mice. The levels of melatonin, cortisol, and the circadian rhythm of Brain and muscle ARNT-like 1 (BMAL1) in PD model mice were detected after optimal frequency rTMS treatment. Additionally, KN-93 and Bmal1siRNA interventions were used to verify that rTMS could alleviate PD symptoms by regulating the CaMKII-CREB-BMAL1 pathway. Results Administration of 10 Hz rTMS significantly improved neurological function, increased the protein levels of TH and BDNF, and inhibited abnormal aggregation of a-syn. Furthermore, administration of 10 Hz rTMS regulated the secretion profile of cortisol and melatonin and reversed the circadian arrhythmia of BMAL1 expression. After the KN-93 intervention, the MPTP+rTMS+KN-93 group exhibited decreased levels of P- Ca2+/calmodulin-dependent protein kinase II (CaMKII)/CaMKII, P-cAMP-response-element-binding protein (CREB)/CREB, BMALI, and TH. After Bmal1siRNA intervention, the protein levels of BMAL1 and TH were significantly reduced in the MPTP+10 Hz+ Bmal1siRNA group. At the same time, there were no significant changes in the proportions of P-CaMKIIα/CaMKIIα and P-CREB/CREB expression levels. Finally, immunohistochemical analysis showed that the number of TH-positive neurons was high in the MPTP+10 Hz group, but decreased significantly after KN-93 and Bmal1siRNA interventions. Conclusion Treatment with 10 Hz rTMS alleviated MPTP-induced PD symptoms by regulating the CaMKII-CREB-BMAL1 pathway. This study provides a comprehensive perspective of the therapeutic mechanisms of rTMS in PD.
Collapse
Affiliation(s)
- Dongdong Chen
- Department of Neurosurgery& Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
- Department of Neurosurgery, The Affiliated Hospital of Jiang Nan University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Surong Qian
- Department of Rehabilitation Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital Rehabilitation Medical Center, Gusu School, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Wenjun Qian
- Department of Rehabilitation Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital Rehabilitation Medical Center, Gusu School, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Miao Wu
- Department of Rehabilitation Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital Rehabilitation Medical Center, Gusu School, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Xinlong Wang
- Department of Rehabilitation Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital Rehabilitation Medical Center, Gusu School, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Haitao Shen
- Department of Neurosurgery& Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Xianming Long
- Department of Rheumatology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Ming Ye
- Department of Neurosurgery& Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Yan Gong
- Department of Rehabilitation Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital Rehabilitation Medical Center, Gusu School, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Gang Chen
- Department of Neurosurgery& Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| |
Collapse
|
3
|
Mazzotta GM, Conte C. Alpha Synuclein Toxicity and Non-Motor Parkinson's. Cells 2024; 13:1265. [PMID: 39120295 PMCID: PMC11311369 DOI: 10.3390/cells13151265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a common multisystem neurodegenerative disorder affecting 1% of the population over the age of 60 years. The main neuropathological features of PD are the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the presence of alpha synuclein (αSyn)-rich Lewy bodies both manifesting with classical motor signs. αSyn has emerged as a key protein in PD pathology as it can spread through synaptic networks to reach several anatomical regions of the body contributing to the appearance of non-motor symptoms (NMS) considered prevalent among individuals prior to PD diagnosis and persisting throughout the patient's life. NMS mainly includes loss of taste and smell, constipation, psychiatric disorders, dementia, impaired rapid eye movement (REM) sleep, urogenital dysfunction, and cardiovascular impairment. This review summarizes the more recent findings on the impact of αSyn deposits on several prodromal NMS and emphasizes the importance of early detection of αSyn toxic species in biofluids and peripheral biopsies as prospective biomarkers in PD.
Collapse
Affiliation(s)
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| |
Collapse
|
4
|
Silva RH, Lopes-Silva LB, Cunha DG, Becegato M, Ribeiro AM, Santos JR. Animal Approaches to Studying Risk Factors for Parkinson's Disease: A Narrative Review. Brain Sci 2024; 14:156. [PMID: 38391730 PMCID: PMC10887213 DOI: 10.3390/brainsci14020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Despite recent efforts to search for biomarkers for the pre-symptomatic diagnosis of Parkinson's disease (PD), the presence of risk factors, prodromal signs, and family history still support the classification of individuals at risk for this disease. Human epidemiological studies are useful in this search but fail to provide causality. The study of well-known risk factors for PD in animal models can help elucidate mechanisms related to the disease's etiology and contribute to future prevention or treatment approaches. This narrative review aims to discuss animal studies that investigated four of the main risk factors and/or prodromal signs related to PD: advanced age, male sex, sleep alterations, and depression. Different databases were used to search the studies, which were included based on their relevance to the topic. Although still in a reduced number, such studies are of great relevance in the search for evidence that leads to a possible early diagnosis and improvements in methods of prevention and treatment.
Collapse
Affiliation(s)
- R H Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo 04021-001, SP, Brazil
| | - L B Lopes-Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo 04021-001, SP, Brazil
| | - D G Cunha
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo 04021-001, SP, Brazil
| | - M Becegato
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo 04021-001, SP, Brazil
| | - A M Ribeiro
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Universidade Federal de São Paulo, Santos 11015-020, SP, Brazil
| | - J R Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana 49500-000, SE, Brazil
| |
Collapse
|
5
|
Chen YC, Wang WS, Lewis SJG, Wu SL. Fighting Against the Clock: Circadian Disruption and Parkinson's Disease. J Mov Disord 2024; 17:1-14. [PMID: 37989149 PMCID: PMC10846969 DOI: 10.14802/jmd.23216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023] Open
Abstract
Circadian disruption is being increasingly recognized as a critical factor in the development and progression of Parkinson's disease (PD). This review aims to provide an in-depth overview of the relationship between circadian disruption and PD by exploring the molecular, cellular, and behavioral aspects of this interaction. This review will include a comprehensive understanding of how the clock gene system and transcription-translation feedback loops function and how they are diminished in PD. The article also discusses the role of clock genes in the regulation of circadian rhythms, as well as the impact of clock gene dysregulation on mitochondrial function, oxidative stress, and neuroinflammation, including the microbiota-gut-brain axis, which have all been proposed as being crucial mechanisms in the pathophysiology of PD. Finally, this review highlights potential therapeutic strategies targeting the clock gene system and circadian rhythm for the treatment of PD.
Collapse
Affiliation(s)
- Yen-Chung Chen
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Sheng Wang
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Simon J G Lewis
- Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Shey-Lin Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Electrical Engineering, National Changhua University of Education, Changhua, Taiwan
| |
Collapse
|
6
|
Townsend LTJ, Anderson KN, Boeve BF, McKeith I, Taylor JP. Sleep disorders in Lewy body dementia: Mechanisms, clinical relevance, and unanswered questions. Alzheimers Dement 2023; 19:5264-5283. [PMID: 37392199 DOI: 10.1002/alz.13350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 07/03/2023]
Abstract
In Lewy body dementia (LBD), disturbances of sleep and/or arousal including insomnia, excessive daytime sleepiness, rapid eye movement (REM) sleep behavior disorder, obstructive sleep apnea, and restless leg syndrome are common. These disorders can each exert a significant negative impact on both patient and caregiver quality of life; however, their etiology is poorly understood. Little guidance is available for assessing and managing sleep disorders in LBD, and they remain under-diagnosed and under-treated. This review aims to (1) describe the specific sleep disorders which occur in LBD, considering their putative or potential mechanisms; (2) describe the history and diagnostic process for these disorders in LBD; and (3) summarize current evidence for their management in LBD and consider some of the ongoing and unanswered questions in this field and future research directions.
Collapse
Affiliation(s)
- Leigh T J Townsend
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
- Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Kirstie N Anderson
- Regional Sleep Service, Newcastle-upon-Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian McKeith
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - John-Paul Taylor
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
- Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
7
|
Chen Y, Gao Y, Sun X, Wang BH, Qin L, Wu IX, Li G. Association between Sleep Factors and Parkinson's Disease: A Prospective Study Based on 409,923 UK Biobank Participants. Neuroepidemiology 2023; 57:293-303. [PMID: 37231899 DOI: 10.1159/000530982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
INTRODUCTION Limited evidence indicates an association between sleep factors and the risk of Parkinson's disease (PD). However, large prospective cohort studies including both sexes are needed to verify the association between daytime sleepiness, sleep duration, and PD risk. Furthermore, other sleep factors like chronotype and snoring and their impact on increased PD risk should be explored by simultaneously considering daytime sleepiness and snoring. METHODS This study included 409,923 participants from the UK Biobank. Data on five sleep factors (chronotype, sleep duration, sleeplessness/insomnia, snoring, and daytime sleepiness) were collected using a standard self-administered questionnaire. PD occurrence was identified using linkages with primary care, hospital admission, death register, or self-report. Cox proportional hazard models were used to investigate the association between sleep factors and PD risk. Subgroup (age and sex) and sensitivity analyses were performed. RESULTS During a median follow-up of 11.89 years, 2,158 incident PD cases were documented. The main association analysis showed that prolonged sleep duration (hazard ratio [HR]: 1.20, 95% confidence interval [CI]: 1.05, 1.37) and occasional daytime sleepiness (HR: 1.15, 95% CI: 1.04, 1.26) increased the PD risk. Compared to those who self-reported never or rarely having sleeplessness/insomnia, participants who reported usually having sleeplessness/insomnia had a decreased risk of PD (HR: 0.85, 95% CI: 0.75, 0.96). Subgroup analysis revealed that women who self-reported no snoring had a decreased PD risk (HR: 0.85; 95% CI: 0.73, 0.99). Sensitivity analyses indicated that the robustness of the results was affected by potential reverse causation and data completeness. CONCLUSION Long sleep duration increased the PD risk, especially among men and participants ≥60 years, while snoring increased the risk of PD in women. Additional studies are needed to (i) further consider other sleep traits (e.g., rapid eye movement sleep behavior disorder and sleep apnea) that might be related to PD, (ii) objectively measure sleep-related exposure, and (iii) confirm the effects of snoring on PD risk by considering the impact of obstructive sleep apnea and investigating its underlying mechanisms.
Collapse
Affiliation(s)
- Yancong Chen
- Changsha Center for Disease Control and Prevention, Changsha, China,
- Xiangya School of Public Health, Central South University, Changsha, China,
| | - Yinyan Gao
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Xuemei Sun
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Betty Huan Wang
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Lang Qin
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Irene Xy Wu
- Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, China
| | - Guowei Li
- MMed, MBBS, CCEM, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
8
|
Cheng WY, Ho YS, Chang RCC. Linking circadian rhythms to microbiome-gut-brain axis in aging-associated neurodegenerative diseases. Ageing Res Rev 2022; 78:101620. [PMID: 35405323 DOI: 10.1016/j.arr.2022.101620] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/13/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022]
Abstract
Emerging evidence suggests that both disruption of circadian rhythms and gut dysbiosis are closely related to aging-associated neurodegenerative diseases. Over the last decade, the microbiota-gut-brain axis has been an emerging field and revolutionized studies in pathology, diagnosis, and treatment of neurological disorders. Crosstalk between the brain and gut microbiota can be accomplished via the endocrine, immune, and nervous system. Recent studies have shown that the composition and diurnal oscillation of gut microbiota are influenced by host circadian rhythms. This provides a new perspective for investigating the microbiome-gut-brain axis. We aim to review current understanding and research on the dynamic interaction between circadian rhythms and the microbiome-gut-brain axis. Furthermore, we will address the possible neurodegenerative disease contribution through circadian rhythms and microbiome-gut-brain axis crosstalk.
Collapse
Affiliation(s)
- Wai-Yin Cheng
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Yuen-Shan Ho
- School of Nursing, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region.
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
9
|
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
|
10
|
He G, Liu CF, Ye Q, Liu Z, Jin M, Shang H, Chen L, Tuo H, Jiang H, Cai J, Joshi K, Cooper J, Zi L, Chen S. Prevalence and profile of nocturnal disturbances in Chinese patients with advanced-stage Parkinson's disease: a cross-sectional epidemiology study. BMC Neurol 2021; 21:194. [PMID: 33980158 PMCID: PMC8114718 DOI: 10.1186/s12883-021-02217-5] [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: 12/03/2020] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Background The impact of nocturnal disturbance (ND) in Parkinson’s disease on quality of life of patients in Western Countries is increasingly understood. Our study aimed to investigate ND prevalence and its quality of life impact in patients with advanced Parkinson’s disease in China. Methods In a multicenter, tertiary-care hospital, outpatient-based, cross-sectional study, patients with advanced Parkinson’s disease (Modified Hoehn & Yahr [H&Y] Stage II–IV with ≥3 h awake “off” time/day) from 10 tertiary hospitals throughout China completed the Parkinson’s Disease Sleep Scale-2 (PDSS-2) and Parkinson’s Disease Questionnaire-39 (PDQ-39). The primary endpoint was the percentage of patients with significant ND (PDSS-2 total score ≥ 15). Additional endpoints were demographic and clinical characteristics, PDSS-2 and PDQ-39 total and subscale scores, correlation between PDSS-2 and PDQ-39, and risk factors for ND and higher PDSS-2 or PDQ-39 scores. Results Of 448 patients analyzed (mean age 63.5 years, 47.3% female), 70.92% (95% confidence interval: 66.71, 75.13) had significant ND. Presence of ND and higher PDSS-2 scores were associated with longer disease duration and higher H&Y stage. Presence of ND was also associated with more awake “off” time/day and female sex. PDQ-39 scores were significantly worse for patients with ND versus those without ND; worse scores were associated with more awake “off” time/day, female sex, and higher H&Y stage. PDSS-2 and PDQ-39 total scores were associated: Pearson correlation coefficient 0.62 (p < 0.001). Conclusions In China, ND was highly prevalent in patients with advanced Parkinson’s disease and adversely impacted quality of life. This study highlights the importance of early diagnosis and optimized management of ND in patients with Parkinson’s disease in China. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02217-5.
Collapse
Affiliation(s)
- Guiying He
- Department of Neurology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Feng Liu
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qinyong Ye
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhenguo Liu
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Jin
- China-Japan Friendship Hospital, Beijing, China
| | - Huifang Shang
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Chen
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Houzhen Tuo
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong Jiang
- Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jifu Cai
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | | | | | | | - Shengdi Chen
- Department of Neurology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Verugina NI, Levin OS, Lyashenko EA. [Neuroendocrine and metabolic impairments in patients with Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:67-73. [PMID: 33205933 DOI: 10.17116/jnevro202012010267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
ABSRACT Neuroendocrine and neurometabolic disorders, although occasionally noted in Parkinson's disease (PD), existed in the shadow of motor and non-motor symptoms (hypokinesia, rigidity, tremor, depression, constipation, etc.). In recent years, they are increasingly being diagnosed and are the subject of special research. These include, in particular, disorders of carbohydrate metabolism, changes in body weight, metabolic disorders in bone tissue, secretion, as well as the secretion of neurohormones, such as melatonin. They are associated with other non-motor symptoms, negatively affect patients' general condition and quality of life, but can be treatable. At the same time, treatment of neuroendocrine and neurometabolic disorders can favorably influence the rate of progression of the disease as a whole. This review discusses the pathophysiological mechanisms, clinical consequences, as well as pharmacological and non-pharmacological approaches to the treatment of neuroendocrine and neurometabolic disorders arising in PD, which have been relatively rarely covered in literature.
Collapse
Affiliation(s)
- N I Verugina
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - O S Levin
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - E A Lyashenko
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
12
|
Requejo C, López-de-Ipiña K, Ruiz-Ortega JÁ, Fernández E, Calvo PM, Morera-Herreras T, Miguelez C, Cardona-Grifoll L, Cepeda H, Ugedo L, Lafuente JV. Changes in Day/Night Activity in the 6-OHDA-Induced Experimental Model of Parkinson's Disease: Exploring Prodromal Biomarkers. Front Neurosci 2020; 14:590029. [PMID: 33154717 PMCID: PMC7591774 DOI: 10.3389/fnins.2020.590029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
The search for experimental models mimicking an early stage of Parkinson's disease (PD) before motor manifestations is fundamental in order to explore early signs and get a better prognosis. Interestingly, our previous studies have indicated that 6-hydroxydopamine (6-OHDA) is a suitable model to induce an early degeneration of the nigrostriatal system without any gross motor impairment. Considering our previous findings, we aim to implement a novel system to monitor rats after intrastriatal injection of 6-OHDA to detect and analyze physiological changes underlying prodromal PD. Twenty male Sprague-Dawley rats were unilaterally injected with 6-OHDA (n = 10) or saline solution (n = 10) into the right striatum and placed in enriched environment cages where the activity was monitored. After 2 weeks, the amphetamine test was performed before the sacrifice. Immunohistochemistry was developed for the morphological evaluation and western blot analysis to assess molecular changes. Home-cage monitoring revealed behavioral changes in response to 6-OHDA administration including significant hyperactivity and hypoactivity during the light and dark phase, respectively, turning out in a change of the circadian timing. A preclinical stage of PD was functionally confirmed with the amphetamine test. Moreover, the loss of tyrosine hydroxylase expression was significantly correlated with the motor results, and 6-OHDA induced early proapoptotic events. Our findings provide evidence for a novel prodromal 6-OHDA model following a customized monitoring system that could give insights to detect non-motor deficits and molecular targets to test neuroprotective/neurorestorative agents.
Collapse
Affiliation(s)
- Catalina Requejo
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neurology, Icahn School of Medicine at Mount Sinai, The Friedman Brain Institute, New York, NY, United States
| | - Karmele López-de-Ipiña
- EleKin Research Group, Department of Systems Engineering and Automation, University of the Basque Country (UPV/EHU), Donostia, Spain
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - José Ángel Ruiz-Ortega
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Elsa Fernández
- EleKin Research Group, Department of Systems Engineering and Automation, University of the Basque Country (UPV/EHU), Donostia, Spain
| | - Pilar M. Calvo
- EleKin Research Group, Department of Systems Engineering and Automation, University of the Basque Country (UPV/EHU), Donostia, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Cristina Miguelez
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Laura Cardona-Grifoll
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Hodei Cepeda
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Luisa Ugedo
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
13
|
Li L, Zhao Z, Ma J, Zheng J, Huang S, Hu S, Gu Q, Chen S. Elevated Plasma Melatonin Levels Are Correlated With the Non-motor Symptoms in Parkinson's Disease: A Cross-Sectional Study. Front Neurosci 2020; 14:505. [PMID: 32508583 PMCID: PMC7248560 DOI: 10.3389/fnins.2020.00505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/22/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Melatonin is the major hormone produced and secreted at night by the pineal gland into the cerebrospinal fluid (CSF) and circulation. The relationship between plasma melatonin levels and Parkinson's disease is not clear. The aim of the current study was to assess plasma melatonin levels in Parkinson's disease (PD) patients and to analysis the relationship between plasma melatonin levels and non-motor symptoms. PARTICIPANTS AND METHODS In this cross-sectional study, we evaluated 61 patients with idiopathic PD [males n = 30 (49.2%), average age 62.4 years (range: 46-73 years)] and a total of 58 healthy volunteers [males n = 30 (51.7%), average age 64.3 years (range: 45-70 years)] who participated in the study. Plasma melatonin levels were measured using an enzyme-linked immunosorbent assay. The severity of disease in PD patients was scored by the Unified Parkinson's Disease Rating Scale and the Hoehn and Yahr Staging scale. The quality of life in PD patients was assessed by the 39-item Parkinson's Disease Questionnaire. The non-motor symptoms were assessed by the 14-item Hamilton Anxiety Rating Scale, the 24-item Hamilton Depression Rating Scale, the Parkinson Disease Sleep Scale, the Epworth Sleepiness Scale and the Non-Motor Symptoms Scale for PD. RESULTS Compared with the healthy controls, the plasma melatonin levels were significantly higher in PD patients (12.82 ± 4.85 vs. 19.40 ± 4.23, P < 0.001). Plasma melatonin levels were significantly associated with the levodopa equivalent daily dose (r = -0.262, P < 0.05, n = 61). Higher plasma melatonin concentrations were detected in the negative cardiovascular symptom group than in the cardiovascular symptom group (20.13 ± 3.74 vs. 16.93 ± 3.74, P < 0.05). Higher plasma melatonin concentrations were detected in the non-sleep-disorders group than in the sleep disorders group (22.12 ± 5.93 vs. 18.86 ± 3.66, P < 0.05). In addition, the plasma melatonin concentration was higher in the group without gastrointestinal dysfunction than in the gastrointestinal dysfunction group (21.71 ± 4.44 vs. 18.35 ± 3.74, P < 0.05). CONCLUSION This study revealed that the plasma melatonin levels in PD patients were significantly higher than those in healthy controls. Non-motor symptoms that were significantly negatively correlated with plasma melatonin levels were cardiovascular symptoms, sleep disorders, and gastrointestinal dysfunction. Plasma melatonin levels have the closest relationship with sleep disorders. There was a correlation between plasma melatonin levels and sleep quality in patients with PD. The remaining non-motor symptoms were not related to plasma melatonin levels.
Collapse
Affiliation(s)
- Linyi Li
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Zhenxiang Zhao
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Jianjun Ma
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Jinhua Zheng
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Shen Huang
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Shiyu Hu
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Henan University, Zhengzhou, China
| | - Qi Gu
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Siyuan Chen
- Department of Neurology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
14
|
Liu WW, Wei SZ, Huang GD, Liu LB, Gu C, Shen Y, Wang XH, Xia ST, Xie AM, Hu LF, Wang F, Liu CF. BMAL1 regulation of microglia-mediated neuroinflammation in MPTP-induced Parkinson's disease mouse model. FASEB J 2020; 34:6570-6581. [PMID: 32246801 DOI: 10.1096/fj.201901565rr] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/21/2019] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
Dysfunction of the circadian rhythm is one of most common nonmotor symptoms in Parkinson's disease (PD), but the molecular role of the circadian rhythm in PD is unclear. We here showed that inactivation of brain and muscle ARNT-like 1 (BMAL1) in 1-methyl-4-phenyl-1,2,4,5-tetrahydropyridine (MPTP)-treated mice resulted in obvious motor functional deficit, loss of dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNpc), decrease of dopamine (DA) transmitter, and increased activation of microglia and astrocytes in the striatum. Time on the rotarod or calorie consumption, and food and water intake were reduced in the Bmal1-/- mice after MPTP treatment, suggesting that absence of Bmal1 may exacerbate circadian and PD motor function. We observed a significant reduction of DANs (~35%) in the SNpc, the tyrosine hydroxylase protein level in the striatum (~60%), the DA (~22%), and 3,4-dihydroxyphenylacetic acid content (~29%), respectively, in MPTP-treated Bmal1-/- mice. Loss of Bmal1 aggravated the inflammatory reaction both in vivo and in vitro. These findings suggest that BMAL1 may play an essential role in the survival of DANs and maintain normal function of the DA signaling pathway via regulating microglia-mediated neuroinflammation in the brain.
Collapse
Affiliation(s)
- Wen-Wen Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Shi-Zhuang Wei
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Guo-Dong Huang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Lu-Bing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chao Gu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yun Shen
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xian-Hui Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, China
| | - Shu-Ting Xia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - An-Mu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li-Fang Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
15
|
Arnao V, Cinturino A, Mastrilli S, Buttà C, Maida C, Tuttolomondo A, Aridon P, D'Amelio M. Impaired circadian heart rate variability in Parkinson's disease: a time-domain analysis in ambulatory setting. BMC Neurol 2020; 20:152. [PMID: 32326894 PMCID: PMC7181578 DOI: 10.1186/s12883-020-01722-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 04/12/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Heart rate variability (HRV) decreases in Parkinson's disease (PD) and it can be considered a marker for cardiovascular dysautonomia. The purpose of this pilot study is to evaluate long-term time-domain analysis of HRV of PD patients and compare the results with those of matched healthy individuals. METHODS Idiopathic PD patients without comorbidity impairing HRV, and age-matched healthy individuals were recruited in a pilot study. A long-term time domain analysis of HRV using 24-h ambulatory ECG was performed. RESULTS Overall, 18 PD patients fulfilling inclusion criteria completed the evaluation (mean age was 55.6 ± 8.8, disease duration: 5.0 ± 4.7). Mean SCOPA-AUT score was 10.1 ± 7.3. Patients were on Hoehn & Yahr stage 1-2 and mean Levodopa Equivalent Dose (LED) was 311 ± 239.9. Mean of the 5-min standard deviation (SD) of R-R intervals distribution (SDNN) for all 5 min segments of the entire recording (ISDNN) was significantly lower in patients compared to controls. ISDNN was significantly different between Parkinson's disease patients and healthy controls. CONCLUSIONS In our population characterized by mild to moderate disease severity, time-domain assessment of HRV seemed to be a potential tool to characterize cardiovascular dysautonomia. Decrease of ISDNN in PD may reflect an autonomic derangement extending all day and night long.
Collapse
Affiliation(s)
- V Arnao
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, 90129, Palermo, Italy
| | - A Cinturino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, 90129, Palermo, Italy
| | - S Mastrilli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, 90129, Palermo, Italy
| | - C Buttà
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), Università degli Studi di Palermo, Palermo, Italy
| | - C Maida
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), Università degli Studi di Palermo, Palermo, Italy
| | - A Tuttolomondo
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), Università degli Studi di Palermo, Palermo, Italy
| | - P Aridon
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, 90129, Palermo, Italy
| | - M D'Amelio
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via Gaetano la Loggia n.1, 90129, Palermo, Italy.
| |
Collapse
|
16
|
Chronic sleep restriction in the rotenone Parkinson's disease model in rats reveals peripheral early-phase biomarkers. Sci Rep 2019; 9:1898. [PMID: 30760786 PMCID: PMC6374389 DOI: 10.1038/s41598-018-37657-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is a chronic disorder that presents a range of premotor signs, such as sleep disturbances and cognitive decline, which are key non-motor features of the disease. Increasing evidence of a possible association between sleep disruption and the neurodegenerative process suggests that sleep impairment could produce a detectable metabolic signature on the disease. In order to integrate neurocognitive and metabolic parameters, we performed untargeted and targeted metabolic profiling of the rotenone PD model in a chronic sleep restriction (SR) (6 h/day for 21 days) condition. We found that SR combined with PD altered several behavioural (reversal of locomotor activity impairment; cognitive impairment; delay of rest-activity rhythm) and metabolic parameters (branched-chain amino acids, tryptophan pathway, phenylalanine, and lipoproteins, pointing to mitochondrial impairment). If combined, our results bring a plethora of parameters that represents reliable early-phase PD biomarkers which can easily be measured and could be translated to human studies.
Collapse
|
17
|
Low-Grade Inflammation Aggravates Rotenone Neurotoxicity and Disrupts Circadian Clock Gene Expression in Rats. Neurotox Res 2018; 35:421-431. [PMID: 30328585 DOI: 10.1007/s12640-018-9968-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/08/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
A single injection of LPS produced low-grade neuroinflammation leading to Parkinson's disease (PD) in mice several months later. Whether such a phenomenon occurs in rats and whether such low-grade neuroinflammation would aggravate rotenone (ROT) neurotoxicity and disrupts circadian clock gene/protein expressions were examined in this study. Male rats were given two injections of LPS (2.5-7.5 mg/kg), and neuroinflammation and dopamine neuron loss were evident 3 months later. Seven months after a single LPS (5 mg/kg) injection, rats received low doses of ROT (0.5 mg/kg, sc, 5 times/week for 4 weeks) to examine low-grade neuroinflammation on ROT toxicity. LPS plus ROT produced more pronounced non-motor and motor dysfunctions than LPS or ROT alone in behavioral tests, and decreased mitochondrial complex 1 activity, together with aggravated neuroinflammation and neuron loss. The expressions of clock core genes brain and muscle Arnt-like protein-1 (Bmal1), locomotor output cycles kaput (Clock), and neuronal PAS domain protein-2 (Npas2) were decreased in LPS, ROT, and LPS plus ROT groups. The expressions of circadian feedback genes Periods (Per1 and Per2) were also decreased, but Cryptochromes (Cry1 and Cry2) were unaltered. The circadian clock target genes nuclear receptor Rev-Erbα (Nr1d1), and D-box-binding protein (Dbp) expressions were also decreased. Consistent with the transcript levels, circadian clock protein BMAL1, CLOCK, NR1D1, and DBP were also decreased. Thus, LPS-induced chronic low-grade neuroinflammation potentiated ROT neurotoxicity and disrupted circadian clock gene/protein expression, suggesting a role of disrupted circadian in PD development and progression. Graphical Abstract ᅟ.
Collapse
|
18
|
Leng Y, Goldman SM, Cawthon PM, Stone KL, Ancoli-Israel S, Yaffe K. Excessive daytime sleepiness, objective napping and 11-year risk of Parkinson's disease in older men. Int J Epidemiol 2018; 47:1679-1686. [PMID: 29873737 PMCID: PMC6208269 DOI: 10.1093/ije/dyy098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2018] [Indexed: 12/26/2022] Open
Abstract
Background It is unknown whether subjective daytime sleepiness or objective napping could precede the risk of Parkinson's disease (PD) in the long term. Methods We studied 2920 men (mean age 76 years) without a history of PD and followed them for 11 years. Excessive daytime sleepiness (EDS) was defined as having an Epworth Sleepiness Scale score >10. Objective naps were defined as ≥5 consecutive minutes of inactivity as measured by actigraphy, and napping duration was the accumulated time of naps outside the main sleep period. We used logistic regression to compare PD risk across four groups: no EDS& napping <1 h/day (N = 1739, 59.5%; referent group), EDS& napping <1 h/day (N = 215, 7.4%), no EDS& napping ≥ 1 h/day (N = 819, 28.1%) and EDS& napping ≥ 1 h/day (N = 147, 5.0%). Results We identified 106 incident PD cases over 11 years. After multivariable adjustment, men with napping ≥ 1h/day alone were twice as likely [odds ratio (OR) = 1.96, 95% confidence interval (CI) 1.25-3.08], and men with both EDS and napping ≥ 1 h/day were almost three times as likely to develop PD (2.52, 1.21-5.27), compared with the referent group. Compared with those with naps for <30 min, men who napped for ≥1 h/day had more than double the risk of PD. No association was found for EDS alone and PD risk. Further adjustment for chronotype and circadian stability, or excluding PD cases identified within 2 years after napping measurements, showed similar results. Conclusions Objective long napping rather than subjective EDS was prospectively associated with a higher risk of PD in older men. Objective measures of napping might be valuable as a preclinical marker for PD.
Collapse
Affiliation(s)
- Yue Leng
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Samuel M Goldman
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Peggy M Cawthon
- Department of Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - Katie L Stone
- Department of Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | | | - Kristine Yaffe
- Departments of Psychiatry, Neurology, and Epidemiology, University of California, and San Francisco VA Medical Center, San Francisco, CA, USA
| |
Collapse
|
19
|
Ortuño-Lizarán I, Esquiva G, Beach TG, Serrano GE, Adler CH, Lax P, Cuenca N. Degeneration of human photosensitive retinal ganglion cells may explain sleep and circadian rhythms disorders in Parkinson's disease. Acta Neuropathol Commun 2018; 6:90. [PMID: 30201049 PMCID: PMC6130068 DOI: 10.1186/s40478-018-0596-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/03/2018] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease (PD) patients often suffer from non-motor symptoms like sleep dysregulation, mood disturbances or circadian rhythms dysfunction. The melanopsin-containing retinal ganglion cells are involved in the control and regulation of these processes and may be affected in PD, as other retinal and visual implications have been described in the disease. Number and morphology of human melanopsin-containing retinal ganglion cells were evaluated by immunohistochemistry in eyes from donors with PD or control. The Sholl number of intersections, the number of branches, and the number of terminals from the Sholl analysis were significantly reduced in PD melanopsin ganglion cells. Also, the density of these cells significantly decreased in PD compared to controls. Degeneration and impairment of the retinal melanopsin system may affect to sleep and circadian dysfunction reported in PD pathology, and its protection or stimulation may lead to better disease prospect and global quality of life of patients.
Collapse
Affiliation(s)
- Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690, San Vicente del Raspeig, Spain
| | - Gema Esquiva
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690, San Vicente del Raspeig, Spain
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | | | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690, San Vicente del Raspeig, Spain
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690, San Vicente del Raspeig, Spain.
| |
Collapse
|
20
|
Oka H, Nakahara A, Umehara T. Rotigotine Improves Abnormal Circadian Rhythm of Blood Pressure in Parkinson's Disease. Eur Neurol 2018; 79:281-286. [PMID: 29763930 DOI: 10.1159/000489574] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/22/2018] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Cardiovascular autonomic failure is commonly associated with Parkinson's disease (PD), affecting the daily lives of patients. Rotigotine was recently reported not to influence cardiovascular autonomic responses in contrast to other dopaminergic drugs. The effect of rotigotine on daily blood pressure (BP) fluctuations might reflect autonomic failure in patients with PD. METHODS Twenty-five PD patients who were receiving rotigotine and 12 patients not receiving rotigotine were recruited. Systolic BP during the daytime and nighttime was measured by 24-h BP monitoring at an interval of 2 years. The patients were divided into 3 groups according to the BP fluctuation type: dippers (nocturnal fall in BP ≥10%), non-dippers (0-10%), and risers (< 0%). The time course of BP was compared between the patients given rotigotine and those not given rotigotine. RESULTS Among the 25 patients who received rotigotine, the BP type worsened in 2 patients, was unchanged in 16 patients, and improved in 7 patients. Among the 12 patients who were not receiving rotigotine, the BP type worsened in 5 patients, was unchanged in 4 patients, and improved only in 3 patients (p = 0.042). CONCLUSION Rotigotine improves the abnormal circadian rhythm of BP in patients with PD. Rotigotine was suggested to have favorable effects on cardiovascular autonomic responses and circadian rhythm in patients with PD.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is the second most common neurodegenerative disorder. Sleep dysfunction is one of the most common non-motor manifestations of PD that has gained significant interest over the past two decades due to its impact on the daily lives of PD patients, poorly understood mechanisms, and limited treatment options. In this review, we discuss the most common sleep disorders in PD and present recent investigations that have broadened our understanding of the epidemiology, clinical manifestations, diagnosis, and treatment of disturbed sleep and alertness in PD. RESENT FINDINGS The etiology of impaired sleep-wake cycles in PD is multifactorial. Sleep dysfunction in PD encompasses insomnia, REM sleep behavior disorder, sleep-disordered breathing, restless legs syndrome, and circadian dysregulation. Despite the high prevalence of sleep dysfunction in PD, evidence supporting the efficacy of treatment strategies is limited. We are at the opportune time to advance our understanding of sleep dysfunction in PD, which will hopefully lead to mechanisms-driven interventions for better sleep and allow us to approach sleep as a modifiable therapeutic target for other non-motor and motor manifestations in PD.
Collapse
Affiliation(s)
- Aleksandar Videnovic
- Movement Disorders Unit, Massachusetts General Hospital, Boston, MA, 02114, USA. .,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA. .,MGH Neurological Clinical Research Institute, 165 Cambridge Street, Suite 600, Boston, MA, 02446, USA.
| |
Collapse
|
22
|
Mantovani S, Smith SS, Gordon R, O'Sullivan JD. An overview of sleep and circadian dysfunction in Parkinson's disease. J Sleep Res 2018; 27:e12673. [PMID: 29493044 DOI: 10.1111/jsr.12673] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/18/2022]
Abstract
Sleep and circadian alterations are amongst the very first symptoms experienced in Parkinson's disease, and sleep alterations are present in the majority of patients with overt clinical manifestation of Parkinson's disease. However, the magnitude of sleep and circadian dysfunction in Parkinson's disease, and its influence on the pathophysiology of Parkinson's disease remains often unclear and a matter of debate. In particular, the confounding influences of dopaminergic therapy on sleep and circadian dysfunction are a major challenge, and need to be more carefully addressed in clinical studies. The scope of this narrative review is to summarise the current knowledge around both sleep and circadian alterations in Parkinson's disease. We provide an overview on the frequency of excessive daytime sleepiness, insomnia, restless legs, obstructive apnea and nocturia in Parkinson's disease, as well as addressing sleep structure, rapid eye movement sleep behaviour disorder and circadian features in Parkinson's disease. Sleep and circadian disorders have been linked to pathological conditions that are often co-morbid in Parkinson's disease, including cognitive decline, memory impairment and neurodegeneration. Therefore, targeting sleep and circadian alterations could be one of the earliest and most promising opportunities to slow disease progression. We hope that this review will contribute to advance the discussion and inform new research efforts to progress our knowledge in this field.
Collapse
Affiliation(s)
- Susanna Mantovani
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Simon S Smith
- Institute for Social Science Research (ISSR), The University of Queensland, Indooroopilly, Australia
| | - Richard Gordon
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia
| | - John D O'Sullivan
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| |
Collapse
|
23
|
Circadian Regulation of Hippocampal-Dependent Memory: Circuits, Synapses, and Molecular Mechanisms. Neural Plast 2018; 2018:7292540. [PMID: 29593785 PMCID: PMC5822921 DOI: 10.1155/2018/7292540] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Circadian modulation of learning and memory efficiency is an evolutionarily conserved phenomenon, occurring in organisms ranging from invertebrates to higher mammalian species, including humans. While the suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master mammalian pacemaker, recent evidence suggests that forebrain regions, including the hippocampus, exhibit oscillatory capacity. This finding, as well as work on the cellular signaling events that underlie learning and memory, has opened promising new avenues of investigation into the precise cellular, molecular, and circuit-based mechanisms by which clock timing impacts plasticity and cognition. In this review, we examine the complex molecular relationship between clock timing and memory, with a focus on hippocampal-dependent tasks. We evaluate how the dysregulation of circadian timing, both at the level of the SCN and at the level of ancillary forebrain clocks, affects learning and memory. Further, we discuss experimentally validated intracellular signaling pathways (e.g., ERK/MAPK and GSK3β) and potential cellular signaling mechanisms by which the clock affects learning and memory formation. Finally, we examine how long-term potentiation (LTP), a synaptic process critical to the establishment of several forms of memory, is regulated by clock-gated processes.
Collapse
|
24
|
CLOCK 3111T/C Variant Correlates with Motor Fluctuation and Sleep Disorders in Chinese Patients with Parkinson's Disease. PARKINSONS DISEASE 2018. [PMID: 29535854 PMCID: PMC5817304 DOI: 10.1155/2018/4670380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background The clock genes controlling biological rhythm play an important role in the pathophysiology of aging. The purpose of this study was to determine whether there is an association between a variant of the circadian locomotor output cycles kaput (CLOCK) gene and circadian dysfunction of Parkinson's disease (PD). Methods Six hundred and forty-six cases of Parkinson's disease from consecutive outpatients and inpatients ward from our hospital were included in this study. Kompetitive allele-specific PCR was used to determine the frequency distribution of genotypes and alleles. The examinations for the PD group were assessed in person in order to evaluate motor symptoms, cognitive function, sleep, and depression, including the Unified Parkinson's Disease Rating Scale (UPDRS), Mini-Mental State Examination (MMSE), Pittsburgh Sleep Quality Index (PSQI), and 17-item Hamilton Rating Scale for Depression (HAMD-17). Results Motor fluctuation (P < 0.001) and sleep disorders (P=0.007) were significantly different between the two groups. These correlations persisted after adjusting for confounding risk factors by further binary logistic regression analysis, suggesting that the CLOCK 3111T/C variant was associated with motor fluctuation (OR = 1.080, P < 0.001) and a subjective sleep disorder (OR = 1.130, P=0.037). Conclusion The CLOCK 3111T/C variant can be an independent risk factor for motor fluctuation and sleep disorder in Parkinson's disease.
Collapse
|
25
|
Martino JK, Freelance CB, Willis GL. The effect of light exposure on insomnia and nocturnal movement in Parkinson's disease: an open label, retrospective, longitudinal study. Sleep Med 2018. [PMID: 29530365 DOI: 10.1016/j.sleep.2018.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Insomnia, hypersomnia and REM Sleep Behavior Disorder (RSBD) during sleep are major problems for patients suffering from Parkinson's disease (PD) but they are also used to predict its onset. While these secondary symptoms detract from the quality of life in PD patients, few treatment options are available due to limited efficacy or risk of complicating the treatment regimen. Light therapy (LT) has been suggested as a strategy for sleep disorders but it has only been implemented recently for use in PD. An open label, retrospective study was undertaken where PD patients had been undergoing LT, using polychromatic light, for four months to 15 years prior. It was found that 1 h exposure to light, just prior to retiring, significantly improved insomnia and reduced RSBD in as little as one month after commencing LT. In addition, the improvement was maintained as long as LT was continued over a four to six year period. The efficacy of LT in alleviating these sleep related conditions was not compromised by time since diagnosis or age of the patient. These results intimate the value of long term application of non-invasive techniques such as LT for treating sleep disorders in PD and justify further controlled trials on the long term efficacy of LT.
Collapse
Affiliation(s)
- Jessica K Martino
- The Bronowski Institute of Behavioural Neuroscience, The Bronowski Clinic, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria, 3444, Australia; The Cairnmillar Institute, School of Psychology, Counselling and Psychotherapy, 993 Burke Road, Camberwell, Victoria, 3142, Australia
| | - Christopher B Freelance
- The Bronowski Institute of Behavioural Neuroscience, The Bronowski Clinic, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria, 3444, Australia; School of BioSciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Gregory L Willis
- The Bronowski Institute of Behavioural Neuroscience, The Bronowski Clinic, Coliban Medical Centre, 19 Jennings Street, Kyneton, Victoria, 3444, Australia.
| |
Collapse
|
26
|
Stewart J, Bachman G, Cooper C, Liu L, Ancoli-Israel S, Alibiglou L. Circadian dysfunction and fluctuations in gait initiation impairment in Parkinson’s disease. Exp Brain Res 2018; 236:655-664. [DOI: 10.1007/s00221-017-5163-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 12/23/2017] [Indexed: 11/24/2022]
|
27
|
Willis GL, Freelance CB. Emerging preclinical interest concerning the role of circadian function in Parkinson's disease. Brain Res 2017; 1678:203-213. [PMID: 28958865 DOI: 10.1016/j.brainres.2017.09.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/22/2017] [Accepted: 09/24/2017] [Indexed: 02/08/2023]
Abstract
The importance of circadian function in the aetiology, progression and treatment of Parkinson's disease is a topic of increasing interest to the scientific and clinical community. While clinical studies on this theme are relatively new and limited in number there are many preclinical studies which explore possible circadian involvement in Parkinson's disease and speculate as to the mechanism by which clinical benefit can be derived by manipulating the circadian system. The present review explores the sequelae of circadian related studies from a historical perspective and reveals mechanisms that may be involved in the aetiology and progression of the disease. A systematic review of these studies also sets the stage for understanding the basic neuroscientific approaches which have been applied and provides new direction from which circadian function can be explored.
Collapse
Affiliation(s)
- Gregory L Willis
- The Bronowski Institute of Behavioural Neuroscience, Coliban Medical Centre, 19 Jennings Street, Kyneton, Vic 3444, Australia.
| | - Christopher B Freelance
- The Bronowski Institute of Behavioural Neuroscience, Coliban Medical Centre, 19 Jennings Street, Kyneton, Vic 3444, Australia
| |
Collapse
|
28
|
Sabbar M, Dkhissi-Benyahya O, Benazzouz A, Lakhdar-Ghazal N. Circadian Clock Protein Content and Daily Rhythm of Locomotor Activity Are Altered after Chronic Exposure to Lead in Rat. Front Behav Neurosci 2017; 11:178. [PMID: 28970786 PMCID: PMC5609114 DOI: 10.3389/fnbeh.2017.00178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022] Open
Abstract
Lead exposure has been reported to produce many clinical features, including parkinsonism. However, its consequences on the circadian rhythms are still unknown. Here we aimed to examine the circadian rhythms of locomotor activity following lead intoxication and investigate the mechanisms by which lead may induce alterations of circadian rhythms in rats. Male Wistar rats were injected with lead or sodium acetate (10 mg/kg/day, i.p.) during 4 weeks. Both groups were tested in the “open field” to quantify the exploratory activity and in the rotarod to evaluate motor coordination. Then, animals were submitted to continuous 24 h recordings of locomotor activity under 14/10 Light/dark (14/10 LD) cycle and in complete darkness (DD). At the end of experiments, the clock proteins BMAL1, PER1-2, and CRY1-2 were assayed in the suprachiasmatic nucleus (SCN) using immunohistochemistry. We showed that lead significantly reduced the number of crossing in the open field, impaired motor coordination and altered the daily locomotor activity rhythm. When the LD cycle was advanced by 6 h, both groups adjusted their daily locomotor activity to the new LD cycle with high onset variability in lead-intoxicated rats compared to controls. Lead also led to a decrease in the number of immunoreactive cells (ir-) of BMAL1, PER1, and PER2 without affecting the number of ir-CRY1 and ir-CRY2 cells in the SCN. Our data provide strong evidence that lead intoxication disturbs the rhythm of locomotor activity and alters clock proteins expression in the SCN. They contribute to the understanding of the mechanism by which lead induce circadian rhythms disturbances.
Collapse
Affiliation(s)
- Mariam Sabbar
- Équipe de Recherche sur les Rythmes Biologiques, Neurosciences et Environnement, Faculté des Sciences, Université Mohammed VRabat, Morocco
| | - Ouria Dkhissi-Benyahya
- INSERM, Stem Cell and Brain Research Institute U1208, University of Lyon, Université Claude Bernard Lyon 1Lyon, France
| | - Abdelhamid Benazzouz
- Institut des Maladies Neurodégénératives, Univ. de Bordeaux, UMR5293Bordeaux, France.,Centre National de la Recherche Scientifique, Institut des Maladies Neurodégénératives, UMR5293Bordeaux, France
| | - Nouria Lakhdar-Ghazal
- Équipe de Recherche sur les Rythmes Biologiques, Neurosciences et Environnement, Faculté des Sciences, Université Mohammed VRabat, Morocco
| |
Collapse
|
29
|
Weymann KB, Lim MM. Sleep Disturbances in TBI and PTSD and Potential Risk of Neurodegeneration. CURRENT SLEEP MEDICINE REPORTS 2017. [DOI: 10.1007/s40675-017-0077-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Julienne H, Buhl E, Leslie DS, Hodge JJL. Drosophila PINK1 and parkin loss-of-function mutants display a range of non-motor Parkinson's disease phenotypes. Neurobiol Dis 2017; 104:15-23. [PMID: 28435104 PMCID: PMC5469398 DOI: 10.1016/j.nbd.2017.04.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/13/2017] [Accepted: 04/16/2017] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is more commonly associated with its motor symptoms and the related degeneration of dopamine (DA) neurons. However, it is becoming increasingly clear that PD patients also display a wide range of non-motor symptoms, including memory deficits and disruptions of their sleep-wake cycles. These have a large impact on their quality of life, and often precede the onset of motor symptoms, but their etiology is poorly understood. The fruit fly Drosophila has already been successfully used to model PD, and has been used extensively to study relevant non-motor behaviours in other contexts, but little attention has yet been paid to modelling non-motor symptoms of PD in this genetically tractable organism. We examined memory performance and circadian rhythms in flies with loss-of-function mutations in two PD genes: PINK1 and parkin. We found learning and memory abnormalities in both mutant genotypes, as well as a weakening of circadian rhythms that is underpinned by electrophysiological changes in clock neurons. Our study paves the way for further work that may help us understand the mechanisms underlying these neglected aspects of PD, thus identifying new targets for treatments to address these non-motor problems specifically and perhaps even to halt disease progression in its prodromal phase.
Collapse
Affiliation(s)
- Hannah Julienne
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, United Kingdom
| | - Edgar Buhl
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, United Kingdom
| | - David S Leslie
- Department of Mathematics and Statistics, Fylde College, Lancaster University, Lancaster LA1 4YF, United Kingdom
| | - James J L Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, United Kingdom.
| |
Collapse
|
31
|
Lauretti E, Di Meco A, Merali S, Praticò D. Circadian rhythm dysfunction: a novel environmental risk factor for Parkinson's disease. Mol Psychiatry 2017; 22:280-286. [PMID: 27046648 DOI: 10.1038/mp.2016.47] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 01/04/2023]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder. Although rare genetically linked cases of PD have been reported, most incidences are sporadic in nature. Late-onset, sporadic PD is thought to result from the combined effects of genetic and environmental risk factors exposure. Sleep and circadian rhythm disorders are recurrent among PD patients and appear early in the disease. Although some evidence supports a relationship between circadian disruption (CD) and PD, whether this is secondary to the motor symptoms or, indeed, is a factor that contributes to the pathogenesis of the disease remains to be investigated. In the present paper, we studied the direct consequence of chronic CD on the development of the phenotype in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridinen) model of PD. Pre-exposure to CD to mice treated with MPTP resulted in an exacerbation of motor deficit and a significant reduction in the capability of acquiring motor skills. These changes were associated with a greater loss of tyrosine hydroxylase cell content and intense neuroinflammation. Taken together, our findings demonstrate that CD by triggering a robust neuroinflammatory reaction and degeneration of the nigral-dopaminergic neuronal system exacerbates motor deficit. They support the novel hypothesis that circadian rhythm disorder is an environmental risk factor for developing PD.
Collapse
Affiliation(s)
- E Lauretti
- Department of Pharmacology, Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - A Di Meco
- Department of Pharmacology, Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - S Merali
- Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - D Praticò
- Department of Pharmacology, Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
32
|
De Pablo-Fernández E, Breen DP, Bouloux PM, Barker RA, Foltynie T, Warner TT. Neuroendocrine abnormalities in Parkinson's disease. J Neurol Neurosurg Psychiatry 2017; 88:176-185. [PMID: 27799297 DOI: 10.1136/jnnp-2016-314601] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/06/2016] [Accepted: 10/13/2016] [Indexed: 12/20/2022]
Abstract
Neuroendocrine abnormalities are common in Parkinson's disease (PD) and include disruption of melatonin secretion, disturbances of glucose, insulin resistance and bone metabolism, and body weight changes. They have been associated with multiple non-motor symptoms in PD and have important clinical consequences, including therapeutics. Some of the underlying mechanisms have been implicated in the pathogenesis of PD and represent promising targets for the development of disease biomarkers and neuroprotective therapies. In this systems-based review, we describe clinically relevant neuroendocrine abnormalities in Parkinson's disease to highlight their role in overall phenotype. We discuss pathophysiological mechanisms, clinical implications, and pharmacological and non-pharmacological interventions based on the current evidence. We also review recent advances in the field, focusing on the potential targets for development of neuroprotective drugs in Parkinson's disease and suggest future areas for research.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernández
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, London, UK
| | - David P Breen
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Pierre M Bouloux
- Centre for Neuroendocrinology, Royal Free Campus, UCL Institute of Neurology, London, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology, London, UK
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, London, UK
| |
Collapse
|
33
|
Musiek ES, Videnovic A. Sleep and clocks - implications for brain health. Neurobiol Sleep Circadian Rhythms 2017; 2:1-3. [PMID: 31236492 PMCID: PMC6575571 DOI: 10.1016/j.nbscr.2016.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 12/31/2016] [Indexed: 11/15/2022] Open
Affiliation(s)
- Erik S Musiek
- Neurology and Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA.,Movement Disorders Unit, Massachusetts General Hospital, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Aleksandar Videnovic
- Movement Disorders Unit, Massachusetts General Hospital, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Smolensky MH, Hermida RC, Reinberg A, Sackett-Lundeen L, Portaluppi F. Circadian disruption: New clinical perspective of disease pathology and basis for chronotherapeutic intervention. Chronobiol Int 2016; 33:1101-19. [PMID: 27308960 DOI: 10.1080/07420528.2016.1184678] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Biological processes are organized in time as innate rhythms defined by the period (τ), phase (peak [Φ] and trough time), amplitude (A, peak-trough difference) and mean level. The human time structure in its entirety is comprised of ultradian (τ < 20 h), circadian (20 h > τ < 28 h) and infradian (τ > 28 h) bioperiodicities. The circadian time structure (CTS) of human beings, which is more complicated than in lower animals, is orchestrated and staged by a brain central multioscillator system that includes a prominent pacemaker - the suprachiasmatic nuclei of the hypothalamus. Additional pacemaker activities are provided by the pineal hormone melatonin, which circulates during the nighttime, and the left and right cerebral cortices. Under ordinary circumstances this system coordinates the τ and Φ of rhythms driven by subservient peripheral cell, tissue and organ clock networks. Cyclic environmental, feeding and social time cues synchronize the endogenous 24 h clocks and rhythms. Accordingly, processes and functions of the internal environment are integrated in time for maximum biological efficiency, and they are also organized and synchronized in time to the external environment to ensure optimal performance and response to challenge. Artificial light at night (ALAN) exposure can alter the CTS as can night work, which, like rapid transmeridian displacement by air travel, necessitates realignment of the Φ of the multitude of 24 h rhythms. In 2001, Stevens and Rea coined the phrase "circadian disruption" (CD) to label the CTS misalignment induced by ALAN and shift work (SW) as a potential pathologic mechanism of the increased risk for cancer and other medical conditions. Current concerns relating to the effects of ALAN exposure on the CTS motivated us to renew our long-standing interest in the possible role of CD in the etiopathology of common human diseases and patient care. A surprisingly large number of medical conditions involve CD: adrenal insufficiency; nocturia; sleep-time non-dipping and rising blood pressure 24 h patterns (nocturnal hypertension); delayed sleep phase syndrome, non-24 h sleep/wake disorder; recurrent hypersomnia; SW intolerance; delirium; peptic ulcer disease; kidney failure; depression; mania; bipolar disorder; Parkinson's disease; Smith-Magenis syndrome; fatal familial insomnia syndrome; autism spectrum disorder; asthma; byssinosis; cancers; hand, foot and mouth disease; post-operative state; and ICU outcome. Poorly conceived medical interventions, for example nighttime dosing of synthetic corticosteroids and certain β-antagonists and cyclic nocturnal enteral or parenteral nutrition, plus lifestyle habits, including atypical eating times and chronic alcohol consumption, also can be causal of CD. Just as surprisingly are the many proven chronotherapeutic strategies available today to manage the CD of several of these medical conditions. In clinical medicine, CD seems to be a common, yet mostly unrecognized, pathologic mechanism of human disease as are the many effective chronotherapeutic interventions to remedy it.
Collapse
Affiliation(s)
- Michael H Smolensky
- a Department of Biomedical Engineering , Cockrell School of Engineering, The University of Texas at Austin , Austin , TX , USA
| | - Ramon C Hermida
- b Bioengineering and Chronobiology Laboratories , Atlantic Research Center for Information and Communication Technologies (AtlantTIC), University of Vigo , Vigo , Spain
| | - Alain Reinberg
- c Unité de Chronobiologie , Fondation A de Rothschild , Paris , Cedex , France
| | - Linda Sackett-Lundeen
- d American Association for Clinical Chronobiology and Chronotherapeutics, Roseville , MN , USA
| | - Francesco Portaluppi
- e Hypertension Center, University Hospital S. Anna and Department of Medical Sciences , University of Ferrara , Ferrara , Italy
| |
Collapse
|
35
|
French IT, Muthusamy KA. A Review of Sleep and Its Disorders in Patients with Parkinson's Disease in Relation to Various Brain Structures. Front Aging Neurosci 2016; 8:114. [PMID: 27242523 PMCID: PMC4876118 DOI: 10.3389/fnagi.2016.00114] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 04/29/2016] [Indexed: 11/13/2022] Open
Abstract
Sleep is an indispensable normal physiology of the human body fundamental for healthy functioning. It has been observed that Parkinson's disease (PD) not only exhibits motor symptoms, but also non-motor symptoms such as metabolic irregularities, altered olfaction, cardiovascular dysfunction, gastrointestinal complications and especially sleep disorders which is the focus of this review. A good understanding and knowledge of the different brain structures involved and how they function in the development of sleep disorders should be well comprehended in order to treat and alleviate these symptoms and enhance quality of life for PD patients. Therefore it is vital that the normal functioning of the body in relation to sleep is well understood before proceeding on to the pathophysiology of PD correlating to its symptoms. Suitable treatment can then be administered toward enhancing the quality of life of these patients, perhaps even discovering the cause for this disease.
Collapse
Affiliation(s)
- Isobel T French
- Department of Surgery, University Malaya Kuala Lumpur, Malaysia
| | | |
Collapse
|
36
|
Videnovic A, Willis GL. Circadian system - A novel diagnostic and therapeutic target in Parkinson's disease? Mov Disord 2016; 31:260-9. [PMID: 26826022 DOI: 10.1002/mds.26509] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 01/05/2023] Open
Abstract
The circadian system regulates biological rhythmicity in the human body. The role of the circadian system in neurological disorders is a theme that is attracting an increasing amount of interest from the scientific community. This has arisen, in part, from emerging evidence that disorders such as Parkinson's disease (PD) are multifactorial with many features exhibiting diurnal fluctuations, thereby suggestive of circadian involvement. Although the importance of fluctuating motor and nonmotor manifestations in PD have been well acknowledged, the role of the circadian system has received little attention until recently. It is proposed that intervening with circadian function provides a novel research avenue down which new strategies for improving symptomatic treatment and slowing of the progressive degenerative process can be approached to lessen the burden of PD. In this article we review the literature describing existing circadian research in PD and its experimental models.
Collapse
Affiliation(s)
- Aleksandar Videnovic
- Movement Disorders Unit, Massachusetts General Hospital, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory L Willis
- The Bronowski Institute of Behavioural Neuroscience, Kyneton, Victoria, Australia
| |
Collapse
|
37
|
Gu Z, Wang B, Zhang YB, Ding H, Zhang Y, Yu J, Gu M, Chan P, Cai Y. Association of ARNTL and PER1 genes with Parkinson's disease: a case-control study of Han Chinese. Sci Rep 2015; 5:15891. [PMID: 26507264 PMCID: PMC4623766 DOI: 10.1038/srep15891] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 10/05/2015] [Indexed: 11/09/2022] Open
Abstract
Circadian disruptions may result in sleep problems, oxidative stress and an altered inflammatory response. These symptoms may contribute to PD pathogenesis, despite a lack of direct experimental evidence supporting this relationship. Clock genes are essential to drive and maintain circadian rhythm. To elucidate the possible role of circadian disruptions in PD, we investigated 132 tag variants in eight clock genes. We genotyped these tags within 1,394 Chinese cases and 1,342 controls using Illumina GoldenGate chips. We discovered that SNPs in ARNTL (rs900147, P = 3.33 × 10(-5), OR = 0.80) and PER1 (rs2253820, P = 5.30 × 10(-6), OR = 1.31) genes are significantly associated with PD risk. Moreover, the positive association of the ARNTL rs900147 variant was more robust in tremor dominant (TD) (P = 3.44 × 10(-4)) than postural instability and gait difficulty (PIGD) cases (P = 6.06 × 10(-2)). The association of the PER1 rs2253820 variant was more robust in PIGD (P = 5.42 × 10(-5)) than TD cases (P = 4.2 × 10(-2)). Haplotype analysis also showed that ARNTL and PER1 were associated with PD. Imputation analysis identified more SNPs within ARNTL and PER1 associated with PD, some of which may affect gene expression through altering the transcription factor binding site. In summary, our findings suggest that genetic polymorphisms in ARNTL and PER1 genes, as well as circadian disruptions, may contribute to PD pathogenesis.
Collapse
Affiliation(s)
- Zhuqin Gu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
| | - BinBin Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- National Research Institute for Family Planning, Beijing 100081, P.R. China
| | - Yong-Biao Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Hui Ding
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
| | - Yanli Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- National Research Institute for Family Planning, Beijing 100081, P.R. China
| | - Mingliang Gu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Piu Chan
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
| | - Yanning Cai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing 100053, P.R. China
| |
Collapse
|
38
|
Age-related changes in large-conductance calcium-activated potassium channels in mammalian circadian clock neurons. Neurobiol Aging 2015; 36:2176-83. [PMID: 25735218 DOI: 10.1016/j.neurobiolaging.2014.12.040] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/18/2023]
Abstract
Aging impairs the function of the suprachiasmatic nucleus (SCN, the central mammalian clock), leading to a decline in the circadian rhythm of many physiological processes, including sleep-wake rhythms. Recent studies have found evidence of age-related changes in the circadian regulation of potassium currents; these changes presumably lead to a decrease in the SCN's electrical rhythm amplitude. Current through large-conductance Ca(2+)-activated K(+) (BK) channels promote rhythmicity in both SCN neuronal activity and behavior. In many neuron types, changes in BK activity are correlated with changes in intracellular Ca(2+) concentration ([Ca(2+)]i). We performed patch-clamp recordings of SCN neurons in aged mice and observed that the circadian modulation of BK channel activity was lost because of a reduction in BK currents during the night. This reduced current diminished the afterhyperpolarization, depolarized the resting membrane potential, widened the action potential, and increased [Ca(2+)]i. These data suggest that reduced BK current increases [Ca(2+)]i by altering the action potential waveform, possibly contributing to the observed age-related phenotype.
Collapse
|
39
|
Videnovic A, Lazar AS, Barker RA, Overeem S. 'The clocks that time us'--circadian rhythms in neurodegenerative disorders. Nat Rev Neurol 2014; 10:683-93. [PMID: 25385339 PMCID: PMC4344830 DOI: 10.1038/nrneurol.2014.206] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Circadian rhythms are physiological and behavioural cycles generated by an endogenous biological clock, the suprachiasmatic nucleus. The circadian system influences the majority of physiological processes, including sleep-wake homeostasis. Impaired sleep and alertness are common symptoms of neurodegenerative disorders, and circadian dysfunction might exacerbate the disease process. The pathophysiology of sleep-wake disturbances in these disorders remains largely unknown, and is presumably multifactorial. Circadian rhythm dysfunction is often observed in patients with Alzheimer disease, in whom it has a major impact on quality of life and represents one of the most important factors leading to institutionalization of patients. Similarly, sleep and circadian problems represent common nonmotor features of Parkinson disease and Huntington disease. Clinical studies and experiments in animal models of neurodegenerative disorders have revealed the progressive nature of circadian dysfunction throughout the course of neurodegeneration, and suggest strategies for the restoration of circadian rhythmicity involving behavioural and pharmacological interventions that target the sleep-wake cycle. In this Review, we discuss the role of the circadian system in the regulation of the sleep-wake cycle, and outline the implications of disrupted circadian timekeeping in neurodegenerative diseases.
Collapse
Affiliation(s)
- Aleksandar Videnovic
- Neurological Clinical Research Institute, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 165 Cambridge Street Suite 650, Boston, MA 02114, USA
| | - Alpar S Lazar
- University of Cambridge, John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, Forvie Site, Cambridge CB2 2PY, UK
| | - Roger A Barker
- University of Cambridge, John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, Forvie Site, Cambridge CB2 2PY, UK
| | - Sebastiaan Overeem
- Department of Neurology, Radboud University Medical Centre, P.O. Box 9101, Nijmegen 6500 HB, Netherlands
| |
Collapse
|
40
|
dos Santos AB, Kohlmeier KA, Barreto GE. Are Sleep Disturbances Preclinical Markers of Parkinson’s Disease? Neurochem Res 2014; 40:421-7. [DOI: 10.1007/s11064-014-1488-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 10/24/2022]
|
41
|
Bassani TB, Gradowski RW, Zaminelli T, Barbiero JK, Santiago RM, Boschen SL, da Cunha C, Lima MMS, Andreatini R, Vital MABF. Neuroprotective and antidepressant-like effects of melatonin in a rotenone-induced Parkinson's disease model in rats. Brain Res 2014; 1593:95-105. [PMID: 25301688 DOI: 10.1016/j.brainres.2014.09.068] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 09/17/2014] [Accepted: 09/27/2014] [Indexed: 12/21/2022]
Abstract
Parkinson׳s disease (PD) is a neurodegenerative disorder characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Systemic and intranigral exposure to rotenone in rodents reproduces many of the pathological and behavioral features of PD in humans and thus has been used as an animal model of the disease. Melatonin is a neurohormone secreted by the pineal gland, which has several important physiological functions. It has been reported to be neuroprotective in some animal models of PD. The present study investigated the effects of prolonged melatonin treatment in rats previously exposed to rotenone. The animals were intraperitoneally treated for 10 days with rotenone (2.5mg/kg) or its vehicle. 24h later, they were intraperitoneally treated with melatonin (10mg/kg) or its vehicle for 28 days. One day after the last rotenone exposure, the animals exhibited hypolocomotion in the open field test, which spontaneously reversed at the last motor evaluation. We verified that prolonged melatonin treatment after dopaminergic lesion did not alter motor function but produced antidepressant-like effects in the forced swim test, prevented the rotenone-induced reduction of striatal dopamine, and partially prevented tyrosine hydroxylase immunoreactivity loss in the SNpc. Our results indicate that melatonin exerts neuroprotective and antidepressant-like effects in the rotenone model of PD.
Collapse
Affiliation(s)
- Taysa B Bassani
- Pharmacology Department, Federal University of Paraná, Brazil
| | | | - Tiago Zaminelli
- Pharmacology Department, Federal University of Paraná, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Camargo SMR, Vuille-dit-Bille RN, Mariotta L, Ramadan T, Huggel K, Singer D, Götze O, Verrey F. The Molecular Mechanism of Intestinal Levodopa Absorption and Its Possible Implications for the Treatment of Parkinson’s Disease. J Pharmacol Exp Ther 2014; 351:114-23. [DOI: 10.1124/jpet.114.216317] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
43
|
Kinoshita C, Aoyama K, Matsumura N, Kikuchi-Utsumi K, Watabe M, Nakaki T. Rhythmic oscillations of the microRNA miR-96-5p play a neuroprotective role by indirectly regulating glutathione levels. Nat Commun 2014; 5:3823. [PMID: 24804999 PMCID: PMC4024755 DOI: 10.1038/ncomms4823] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 04/07/2014] [Indexed: 12/11/2022] Open
Abstract
Glutathione (GSH) is a key antioxidant that plays an important neuroprotective role in the brain. Decreased GSH levels are associated with neurodegenerative diseases such as Parkinson’s disease and Alzheimer’s disease. Here we show that a diurnal fluctuation of GSH levels is correlated with neuroprotective activity against oxidative stress in dopaminergic cells. In addition, we found that the cysteine transporter excitatory amino acid carrier 1 (EAAC1), which is involved in neuronal GSH synthesis, is negatively regulated by the microRNA miR-96-5p, which exhibits a diurnal rhythm. Blocking miR-96-5p by intracerebroventricular administration of an inhibitor increased the level of EAAC1 as well as that of GSH and had a neuroprotective effect against oxidative stress in the mouse substantia nigra. Our results suggest that the diurnal rhythm of miR-96-5p may play a role in neuroprotection by regulating neuronal GSH levels via EAAC1. Glutathione is a key antioxidant that plays an important neuroprotective role in the brain. Here, Kinoshita et al. show that levels of glutathione exhibit diurnal fluctuations that are indirectly regulated by the microRNA miR-96-5p, and that this microRNA plays a neuroprotective role against oxidative stress.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Nobuko Matsumura
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Kazue Kikuchi-Utsumi
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Masahiko Watabe
- 1] Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan [2] General Medical Education Center (G-MEC), Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Toshio Nakaki
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| |
Collapse
|
44
|
Circadian dysfunction in response to in vivo treatment with the mitochondrial toxin 3-nitropropionic acid. ASN Neuro 2014; 6:e00133. [PMID: 24328694 PMCID: PMC3891360 DOI: 10.1042/an20130042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sleep disorders are common in neurodegenerative diseases including Huntington's disease (HD) and develop early in the disease process. Mitochondrial alterations are believed to play a critical role in the pathophysiology of neurodegenerative diseases. In the present study, we evaluated the circadian system of mice after inhibiting mitochondrial complex II of the respiratory chain with the toxin 3-nitropropionic acid (3-NP). We found that a subset of mice treated with low doses of 3-NP exhibited severe circadian deficit in behavior. The temporal patterning of sleep behavior is also disrupted in some mice with evidence of difficulty in the initiation of sleep behavior. Using the open field test during the normal sleep phase, we found that the 3-NP-treated mice were hyperactive. The molecular clockwork responsible for the generation of circadian rhythms as measured by PER2::LUCIFERASE was disrupted in a subset of mice. Within the SCN, the 3-NP treatment resulted in a reduction in daytime firing rate in the subset of mice which had a behavioral deficit. Anatomically, we confirmed that all of the treated mice showed evidence for cell loss within the striatum but we did not see evidence for gross SCN pathology. Together, the data demonstrates that chronic treatment with low doses of the mitochondrial toxin 3-NP produced circadian deficits in a subset of treated mice. This work does raise the possibility that the neural damage produced by mitochondrial dysfunction can contribute to the sleep/circadian dysfunction seen so commonly in neurodegenerative diseases.
Collapse
|
45
|
Nutrition and Gastrointestinal Health as Modulators of Parkinson’s Disease. PHARMA-NUTRITION 2014. [DOI: 10.1007/978-3-319-06151-1_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
46
|
Grieb B, von Nicolai C, Engler G, Sharott A, Papageorgiou I, Hamel W, Engel AK, Moll CK. Decomposition of abnormal free locomotor behavior in a rat model of Parkinson's disease. Front Syst Neurosci 2013; 7:95. [PMID: 24348346 PMCID: PMC3842038 DOI: 10.3389/fnsys.2013.00095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 11/08/2013] [Indexed: 11/13/2022] Open
Abstract
Poverty of spontaneous movement, slowed execution and reduced amplitudes of movement (akinesia, brady- and hypokinesia) are cardinal motor manifestations of Parkinson's disease that can be modeled in experimental animals by brain lesions affecting midbrain dopaminergic neurons. Most behavioral investigations in experimental parkinsonism have employed short-term observation windows to assess motor impairments. We postulated that an analysis of longer-term free exploratory behavior could provide further insights into the complex fine structure of altered locomotor activity in parkinsonian animals. To this end, we video-monitored 23 h of free locomotor behavior and extracted several behavioral measures before and after the expression of a severe parkinsonian phenotype following bilateral 6-hydroxydopamine (6-OHDA) lesions of the rat dopaminergic substantia nigra. Unbiased stereological cell counting verified the degree of midbrain tyrosine hydroxylase positive cell loss in the substantia nigra and ventral tegmental area. In line with previous reports, overall covered distance and maximal motion speed of lesioned animals were found to be significantly reduced compared to controls. Before lesion surgery, exploratory rat behavior exhibited a bimodal distribution of maximal speed values obtained for single movement episodes, corresponding to a "first" and "second gear" of motion. 6-OHDA injections significantly reduced the incidence of second gear motion episodes and also resulted in an abnormal prolongation of these fast motion events. Likewise, the spatial spread of such episodes was increased in 6-OHDA rats. The increase in curvature of motion tracks was increased in both lesioned and control animals. We conclude that the discrimination of distinct modes of motion by statistical decomposition of longer-term spontaneous locomotion provides useful insights into the fine structure of fluctuating motor functions in a rat analog of Parkinson's disease.
Collapse
Affiliation(s)
- Benjamin Grieb
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany ; Department of General Psychiatry, Center for Psychosocial Medicine, University of Heidelberg Heidelberg, Germany
| | - Constantin von Nicolai
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany ; Centre for Integrative Neuroscience, University of Tübingen Tübingen, Germany
| | - Gerhard Engler
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany
| | - Andrew Sharott
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany ; Medical Research Council, Anatomical Neuropharacology Unit, Department of Pharmacology, University of Oxford Oxford, UK
| | - Ismini Papageorgiou
- Division of General Neurophysiology, Institute of Physiology and Pathophysiology, University of Heidelberg Heidelberg, Germany
| | - Wolfgang Hamel
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany
| | - Andreas K Engel
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany
| | - Christian K Moll
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, University of Hamburg Hamburg, Germany
| |
Collapse
|
47
|
Schroeder AM, Colwell CS. How to fix a broken clock. Trends Pharmacol Sci 2013; 34:605-19. [PMID: 24120229 PMCID: PMC3856231 DOI: 10.1016/j.tips.2013.09.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/10/2013] [Accepted: 09/11/2013] [Indexed: 12/29/2022]
Abstract
Fortunate are those who rise out of bed to greet the morning light well rested with the energy and enthusiasm to drive a productive day. Others, however, depend on hypnotics for sleep and require stimulants to awaken lethargic bodies. Sleep/wake disruption is a common occurrence in healthy individuals throughout their lifespan and is also a comorbid condition to many diseases (neurodegenerative) and psychiatric disorders (depression and bipolar). There is growing concern that chronic disruption of the sleep/wake cycle contributes to more serious conditions including diabetes (type 2), cardiovascular disease, and cancer. A poorly functioning circadian system resulting in misalignments in the timing of clocks throughout the body may be at the root of the problem for many people. In this article we discuss environmental (light therapy) and lifestyle changes (scheduled meals, exercise, and sleep) as interventions to help fix a broken clock. We also discuss the challenges and potential for future development of pharmacological treatments to manipulate this key biological system.
Collapse
Affiliation(s)
- Analyne M Schroeder
- Laboratory of Circadian and Sleep Medicine, Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA 90024, USA
| | | |
Collapse
|