1
|
Shentu W, Kong Q, Zhang Y, Li W, Chen Q, Yan S, Wang J, Lai Q, Xu Q, Qiao S. Functional abnormalities of the glymphatic system in cognitive disorders. Neural Regen Res 2025; 20:3430-3447. [PMID: 39820293 PMCID: PMC11974647 DOI: 10.4103/nrr.nrr-d-24-01049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 01/19/2025] Open
Abstract
Various pathological mechanisms represent distinct therapeutic targets for cognitive disorders, but a balance between clearance and production is essential for maintaining the stability of the brain's internal environment. Thus, the glymphatic system may represent a common pathway by which to address cognitive disorders. Using the established model of the glymphatic system as our foundation, this review disentangles and analyzes the components of its clearance mechanism, including the initial inflow of cerebrospinal fluid, the mixing of cerebrospinal fluid with interstitial fluid, and the outflow of the mixed fluid and the clearance. Each section summarizes evidence from experimental animal models and human studies, highlighting the normal physiological properties of key structures alongside their pathological manifestations in cognitive disorders. The same pathologic manifestations of different cognitive disorders appearing in the glymphatic system and the same upstream influences are main points of interest of this review. We conclude this article by discussing new findings and outlining the limitations identified in current research progress.
Collapse
Affiliation(s)
- Wuyue Shentu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qi Kong
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Yier Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Wenyao Li
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qiulu Chen
- Department of Neurology, Zhejiang Medical & Health Group Hangzhou Hospital, Hangzhou, Zhejiang Province, China
| | - Sicheng Yan
- Department of Neurology, Liuzhou People’s Hospital, Liuzhou, Guangxi Zhuang Autonomous Region, China
| | - Junjun Wang
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Qilun Lai
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Qi Xu
- Department of Radiology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Song Qiao
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Ma J, Chen M, Liu GH, Gao M, Chen NH, Toh CH, Hsu JL, Wu KY, Huang CM, Lin CM, Fang JT, Lee SH, Lee TMC. Effects of sleep on the glymphatic functioning and multimodal human brain network affecting memory in older adults. Mol Psychiatry 2025; 30:1717-1729. [PMID: 39397082 PMCID: PMC12014484 DOI: 10.1038/s41380-024-02778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Understanding how sleep affects the glymphatic system and human brain networks is crucial for elucidating the neurophysiological mechanism underpinning aging-related memory declines. We analyzed a multimodal dataset collected through magnetic resonance imaging (MRI) and polysomnographic recording from 72 older adults. A proxy of the glymphatic functioning was obtained from the Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) index. Structural and functional brain networks were constructed based on MRI data, and coupling between the two networks (SC-FC coupling) was also calculated. Correlation analyses revealed that DTI-ALPS was negatively correlated with sleep quality measures [e.g., Pittsburgh Sleep Quality Index (PSQI) and apnea-hypopnea index]. Regarding human brain networks, DTI-ALPS was associated with the strength of both functional connectivity (FC) and structural connectivity (SC) involving regions such as the middle temporal gyrus and parahippocampal gyrus, as well as with the SC-FC coupling of rich-club connections. Furthermore, we found that DTI-ALPS positively mediated the association between sleep quality and rich-club SC-FC coupling. The rich-club SC-FC coupling further mediated the association between DTI-ALPS and memory function in good sleepers but not in poor sleepers. The results suggest a disrupted glymphatic-brain relationship in poor sleepers, which underlies memory decline. Our findings add important evidence that sleep quality affects cognitive health through the underlying neural relationships and the interplay between the glymphatic system and multimodal brain networks.
Collapse
Affiliation(s)
- Junji Ma
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Menglu Chen
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Geng-Hao Liu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mengxia Gao
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Ning-Hung Chen
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Sleep Center, Respiratory Therapy, Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng Hong Toh
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
- Department of Neurology, at Linkou, Chang Gung Memorial Hospital and College of Medicine, Neuroscience Research Center, Chang-Gung University, Taoyuan, Taiwan
- Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yi Wu
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chih-Mao Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chih-Ming Lin
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ji-Tseng Fang
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Shwu-Hua Lee
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China.
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Nenert R, Mueller C, Catiul C, Pilkington J, LeVan P, Sharma A, Szaflarski JP, Amara AW. Brain physiological pulsations are linked to sleep architecture and cognitive performance in older adults. Neuroimage 2025; 311:121187. [PMID: 40187437 DOI: 10.1016/j.neuroimage.2025.121187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The glymphatic system facilitates efficient waste clearance in the brain through the movement of cerebrospinal fluid (CSF) along perivascular spaces. Animal studies have demonstrated that glymphatic efficiency declines with age, but evidence for such decline in humans is limited. We hypothesized that reduced glymphatic efficiency in older adults may be related to age-related worsening of sleep quality, potentially contributing to cognitive impairment. METHODS 20 participants aged ≥60 years provided multi-dimensional cognitive measures, overnight polysomnography, and Magnetic Resonance Encephalography (MREG) performed the morning following the PSG. MREG is a single-shot, three-dimensional (3D) sequence employing a spherical stack-of-spirals trajectory that undersamples 3D k-space, enabling whole-brain data acquisition every 100 milliseconds to non-invasively and dynamically assess brain physiological pulsations. Spectral power and optical flow analyses quantified physiological pulsations within cardiovascular (CvB; 0.52-1.6 Hz), respiratory (RFB; 0.11-0.44 Hz), and low-frequency (LFB; 0.008-0.1 Hz) bands. These measures were correlated with cognitive test scores and sleep parameters assessed by overnight polysomnography. RESULTS Significant associations emerged between physiological pulsations, sleep, and cognitive measures. Cardiovascular pulsation strength correlated with non-rapid eye movement (NREM) stage 3 (N3) sleep percentage (peak voxel in right frontal pole; r = 0.72, p < 0.001) and language domain performance (left calcarine gyrus; r = 0.56, p = 0.01). Respiratory pulsations correlated strongly with sleep onset latency (right inferior temporal gyrus; r = 0.75, p < 0.001). Additionally, low-frequency pulsations were associated with sleep onset latency (right precentral gyrus; r = 0.67, p = 0.002). These findings suggest that glymphatic efficiency, as reflected by brain pulsations, is closely linked to sleep quality and cognitive performance in older adults, particularly involving cortical and subcortical structures relevant to cognitive and sleep regulatory functions. CONCLUSION This study uniquely demonstrates that brain physiological pulsations measured non-invasively with MREG are significantly associated with sleep architecture and cognitive performance in older adults. These findings underscore the potential of MREG to assess glymphatic function and provide important insights into the mechanisms linking sleep disturbances, cognitive decline, and aging. The identified correlations between pulsations and specific brain regions highlight potential pathways through which impaired glymphatic function could contribute to cognitive decline in older adults, suggesting promising avenues for future clinical and research applications.
Collapse
Affiliation(s)
- Rodolphe Nenert
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA.
| | - Christina Mueller
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Corina Catiul
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Jennifer Pilkington
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Pierre LeVan
- Dept. of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Ayushe Sharma
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA
| | - Jerzy P Szaflarski
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA; Neurobiology, USA; Neurosurgery, USA; UAB Epilepsy Center, Birmingham, AL, USA
| | - Amy W Amara
- University of Alabama at Birmingham (UAB) Heersink School of Medicine Departments of Neurology, USA; University of Colorado Anschutz Medical Campus Department of Neurology, Aurora, CO, USA
| |
Collapse
|
4
|
Roura I, Pardo J, Martín-Barceló C, Falcon C, Oltra J, Campabadal A, Bargalló N, Serradell M, Mayà G, Montini A, Pont-Sunyer C, Gaig C, Buongiorno M, Junqué C, Iranzo A, Segura B. Clinical and brain volumetric correlates of decreased DTI-ALPS, suggestive of local glymphatic dysfunction, in iRBD. NPJ Parkinsons Dis 2025; 11:87. [PMID: 40268930 PMCID: PMC12018923 DOI: 10.1038/s41531-025-00942-z] [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: 12/13/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Glymphatic alterations may underlie neurodegeneration in alpha-synucleinopathies. Reduced Diffusion-Tensor Imaging ALong the Perivascular Space (DTI-ALPS), a proxy of perivascular glymphatic activity, has been scarcely studied in isolated REM sleep behaviour disorder (iRBD), a prodromal synucleinopathy stage. Furthermore, its associations with clinical symptoms and brain structural abnormalities remain unexplored. We assessed the DTI-ALPS in sixty-two patients with iRBD and twenty-three healthy controls (HC), exploring its associations with clinical symptoms, cortical thickness and brain volumetric data. iRBD patients exhibited a lower DTI-ALPS and poorer odor identification, semantic fluency and processing speed relative to HC. The DTI-ALPS positively correlated with cognitive performance, olfactory function and amygdalar, hippocampal, brainstem and diencephalic volumes, and negatively with age in iRBD. Perivascular glymphatic activity is compromised in iRBD and is associated with brain atrophy and clinical risk factors of progression to alpha-synucleinopathies, supporting the potential of the DTI-ALPS index as an early imaging neurodegeneration marker.
Collapse
Affiliation(s)
- Ignacio Roura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
| | - Jèssica Pardo
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
| | - Cristina Martín-Barceló
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
| | - Carles Falcon
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Biomedical Imaging Group, Centro de Investigación Biomédica en Red sobre Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Catalonia, Spain
| | - Javier Oltra
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Aging Research Center, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Anna Campabadal
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Neurology Service, Consorci Corporació Sanitària Parc Taulí de Sabadell, Barcelona, Catalonia, Spain
| | - Nuria Bargalló
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Imaging Diagnostic Center (CDI), Hospital Clínic Universitari de Barcelona, Barcelona, Catalonia, Spain
| | - Mònica Serradell
- Sleep Unit, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
| | - Gerard Mayà
- Sleep Unit, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
| | - Angelica Montini
- Sleep Unit, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
| | - Claustre Pont-Sunyer
- Movement Disorders Unit, Neurology Service, Fundació Privada Hospital Asil de, Granollers, Barcelona, Catalonia, Spain
| | - Carles Gaig
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Sleep Unit, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Catalonia, Spain
| | | | - Carme Junqué
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Catalonia, Spain
| | - Alex Iranzo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
- Sleep Unit, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Catalonia, Spain.
| | - Bàrbara Segura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Catalonia, Spain.
| |
Collapse
|
5
|
Wang W, Zhao L, He Z, Zhao Y, Jiang G, Gong C, Zhang Y, Yu J, Liang T, Guo L. Decoding Multifaceted Roles of Sleep-Related Genes as Molecular Bridges in Chronic Disease Pathogenesis. Int J Mol Sci 2025; 26:2872. [PMID: 40243466 PMCID: PMC11988575 DOI: 10.3390/ijms26072872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/01/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Sleep is a fundamental process essential for all organisms. Sleep deprivation can lead to significant detrimental effects, contributing to various physiological disorders and elevating the risk of several diseases. Investigating the relationship between sleep and human diseases offers valuable insights into the molecular mechanisms governing sleep regulation, potentially guiding the development of more effective treatments for sleep disorders and associated diseases. This study explored the roles of sleep-related genes in biological processes and their associations with chronic diseases, mainly including neurological, metabolic, cardiovascular diseases, and cancer. Additionally, an analysis on the sleep-related genes was also performed to understand the potential role in tumorigenesis. This review aims to enhance the understanding of the link between sleep-related genes and chronic diseases, contributing to the development of novel therapeutic approaches targeting sleep and circadian rhythm-related chronic diseases.
Collapse
Affiliation(s)
- Wenyuan Wang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Linjie Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Zhiheng He
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Yang Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Guijie Jiang
- School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| | - Chengjun Gong
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Yan Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Jiafeng Yu
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China;
| | - Tingming Liang
- School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| |
Collapse
|
6
|
Ke S, Luo T, Ding Y, Tang CJ, Jie Z, Shen JZ, Wu D, Du Y. Does Obstructive sleep apnea mediate the risk of cognitive impairment by expanding the perivascular space? Sleep Breath 2025; 29:130. [PMID: 40085157 DOI: 10.1007/s11325-025-03291-6] [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: 11/29/2024] [Revised: 02/08/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a neglected global health issue and when left untreated could lead to cognitive impairment (CI), one of the most burdensome outcomes of OSA. Enlarged perivascular spaces (EPVS), an imaging feature as well as a subtype of cerebral small vessel disease and integral part of CSVD, are associated with cognitive function, but the relationship between EPVS and CI is not well understood and by extension the correlation between OSA and EPVS, how CI develops under the joint impact of OSA and EPVS remains unclear. It is the goal of This study to explore the associations among OSA, EPVS, and CI. METHODS This cross-sectional study included 175 older adults with imaging features of EPVS with or without other CSVD subtype features by cranial magnetic resonance imaging between January 2021 and June 2023 at the Shanghai Fifth People's Hospital. We assessed OSA using polysomnography. Blood samples were collected to determine vascular risk factor indices. Cognitive scoring modalities included the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MOCA). To explore the relationship among OSA, EPVS, and CI, we used single-factor analysis, multifactorial analysis, and receiver operating characteristic (ROC) curves. RESULTS A total of 136 participants were analyzed. In our statistical process, MMSE showed a more distinguished performance than MoCA. Participants with OSA had greater EPVS burdens in the midbrain (p < 0.001) and hippocampus (p < 0.001) and more serious CI (p = 0.001). OSA positively influenced EPVS in the midbrain (β = 0.052; 95% confidence interval [CI]: 0.006, 0.097; p = 0.026) and hippocampus (β = 0.190, 95% CI: 0.104, 0.275, p < 0.001). Moreover, the apnea-hypopnea index (AHI; β = -0.514; 95% CI: -0.077, -0.031; p < 0.001) negatively affected cognitive e function. With each increase in the AHI by 1 unit, the risk of CI increased by 12.0% (odds ratio = 1.120; 95%CI: 1.062, 1.181; p < 0.001). The AHI (sensitivity, 67.20%; specificity, 92.20%; area under the ROC curve, 0.828; p < 0.001) had a certain degree of accuracy in ruling out CI in the EPVS population, as calculated using the ROC curve. CONCLUSIONS We identified significant relations among OSA, EPVS, and CI. The AHI is a potential marker for estimating cognitive function in patients with EPVS.
Collapse
Affiliation(s)
- Shuan Ke
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China
| | - Tianjing Luo
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China
| | - Yi Ding
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China
| | - Chia-Jung Tang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China
| | | | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China.
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China.
| | - Yong Du
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, 200240, China.
| |
Collapse
|
7
|
Kawalec-Rutkowska AM, Czaja J, Skuła M, Simka M. Blood Flow in the Internal Jugular Veins in the Lateral Decubitus Body Position in the Healthy People. J Clin Med 2025; 14:1211. [PMID: 40004742 PMCID: PMC11856538 DOI: 10.3390/jcm14041211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Some studies have suggested that the lateral decubitus position during sleep may protect the brain from neurodegenerative processes. Although the mechanisms of such possible protection are not known, an optimal venous outflow may be responsible. Venous outflow from the cranial cavity is dependent on the body's position. However, to date, flow in the internal jugular veins (IJVs) in the lateral position has not been studied quantitatively. Methods: Using ultrasonography, we measured the cross-sectional areas and flow volumes in the IJVs in a group of 25 healthy individuals aged 20-52 ± 12.1 years. These measurements were performed in the supine, upright, and lateral decubitus positions. Results: In the lateral decubitus position, we revealed a collapse of the IJV located higher, dilatation of the opposite vein, and a shift in flow from one vein to the opposite. In the right lateral position, the mean cross-sectional area and flow in the right IJV were 88.6 ± 71.1 mm2 and 74.3 ± 97.5 mL/min, in the left IJV: 37.2 ± 33.4 mm2 and 48.8 ± 82.8 mL/min. In the left lateral position, the right IJV was 38.4 ± 30.7 mm2 and 56.7 ± 56.1 mL/min, and the left IJV was 75.9 ± 51.9 mm2 and 99.7 ± 123.9 mL/min. However, there was also a high heterogeneity of the cross-sectional area changes, and in many participants, this pattern was not observed. Regarding flow volumes in the lateral body positions, in comparison with the supine position, the total outflow through both internal jugular veins was not significantly different. Conclusions: In terms of venous outflow, the lateral decubitus position did not differ significantly from the supine position. The working hypothesis of a potentially protective effect of this body position during sleep against neurodegeneration through improved venous outflow has not been proven, at least in healthy individuals.
Collapse
Affiliation(s)
| | | | | | - Marian Simka
- Institute of Medical Sciences, University of Opole, 45-060 Opole, Poland; (A.M.K.-R.); (J.C.); (M.S.)
| |
Collapse
|
8
|
Chen S, Wang H, Zhang L, Xi Y, Lu Y, Yu K, Zhu Y, Regina I, Bi Y, Tong F. Glymphatic system: a self-purification circulation in brain. Front Cell Neurosci 2025; 19:1528995. [PMID: 40012567 PMCID: PMC11861344 DOI: 10.3389/fncel.2025.1528995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The glymphatic system theory introduces a new perspective on fluid flow and homeostasis in the brain. Here, cerebrospinal fluid and interstitial fluid (CSF-ISF) moves from the perivascular spaces (PVS) of arteries to those of veins for drainage. Aquaporin-4 (AQP4) plays a crucial role in driving fluid within the PVS. The impairment to AQP4 is closely linked to the dysfunction of the glymphatic system. The function of the glymphatic system is less active during waking but enhanced during sleep. The efficiency of the glymphatic system decreases with aging. Damage to the glymphatic system will give rise to the development and progression of many brain diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), chronic traumatic encephalopathy (CTE), and vascular dementia (VaD). Here, we reviewed previous research associated with the glymphatic system, including its concepts, principles, and influencing factors. We hypothesize that AQP4 could be a target for the prevention and treatment of certain brain diseases through the regulation on the glymphatic system.
Collapse
Affiliation(s)
- Siying Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Huijing Wang
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lini Zhang
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yingying Xi
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yiying Lu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Kailin Yu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yujie Zhu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Izmailova Regina
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yong Bi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Fang Tong
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
9
|
Nepozitek J, Dusek P, Sonka K. Glymphatic system, sleep, and Parkinson's disease: interconnections, research opportunities, and potential for disease modification. Sleep 2025; 48:zsae251. [PMID: 39450429 PMCID: PMC11725518 DOI: 10.1093/sleep/zsae251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Jiri Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| |
Collapse
|
10
|
Fanciulli A, Sixel-Döring F, Buhmann C, Krismer F, Hermann W, Winkler C, Woitalla D, Jost WH, Trenkwalder C, Höglinger G. Diagnosis and treatment of autonomic failure, pain and sleep disturbances in Parkinson's disease: guideline "Parkinson's disease" of the German Society of Neurology. J Neurol 2025; 272:90. [PMID: 39751950 PMCID: PMC11698777 DOI: 10.1007/s00415-024-12730-5] [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: 08/08/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND OBJECTIVE Non-motor symptoms frequently develop throughout the disease course of Parkinson's disease (PD), and pose affected individuals at risk of complications, more rapid disease progression and poorer quality of life. Addressing such symptom burden, the 2023 revised "Parkinson's disease" guideline of the German Society of Neurology aimed at providing evidence-based recommendations for managing PD non-motor symptoms, including autonomic failure, pain and sleep disturbances. METHODS Key PICO (Patient, Intervention, Comparison, Outcome) questions were formulated by the steering committee and refined by the assigned authors. Recommendations were drafted based on relevant studies, systematic reviews, meta-analyses and high-quality guidelines identified by the literature search. They were subsequently reviewed, revised, and voted by the Guideline Group in online consensus conferences. Consensus was achieved in case of > 75% agreement among the group members. The consensus was considered strong, if agreement was > 95%. RESULTS The guideline entails: (i) 10 PICOs and 23 recommendations on the diagnosis and treatment of urogenital, cardiovascular and gastrointestinal autonomic failure; (ii) four PICOs and four recommendations on the possible types of pain in PD individuals, their diagnosis and treatment; (iii) 11 PICOs and 11 recommendations on the screening, diagnosis and treatment of sleep disturbances and excessive daytime sleepiness in PD individuals, as well as on their prognostic implications. Thirty-one out of 38 recommendations achieved a strong consensus. CONCLUSION The current German PD guideline provides a practice-oriented and etiology-driven stepwise approach to the diagnosis and treatment of autonomic failure, pain and sleep disturbances in PD individuals.
Collapse
Affiliation(s)
- Alessandra Fanciulli
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-Universität, Marburg, Germany
| | - Carsten Buhmann
- Department of Neurology, University Clinic Eppendorf, Hamburg, Germany
| | - Florian Krismer
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Wiebke Hermann
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Christian Winkler
- Department of Neurology, Lindenbrunn Hospital, Coppenbrügge, Germany
| | | | | | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Günter Höglinger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
11
|
Antelmi E, Lanza G, Mogavero MP, Mingolla GP, Plazzi G, Ferini‐Strambi L, Ferri R, Tinazzi M. Intersection of Sleep Disorders and Parkinson Disease: Unveiling the Bidirectional Relationship. Mov Disord Clin Pract 2025; 12:11-20. [PMID: 39508600 PMCID: PMC11736876 DOI: 10.1002/mdc3.14254] [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: 07/07/2024] [Revised: 08/20/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Patients with Parkinson's Disease (PD) frequently exhibit non-motor symptoms, particularly sleep disturbances. Sleep disorders in PD patients are intricately linked to the pathogenesis and progression of PD itself, exacerbating neurodegenerative processes and worsening patient quality of life. OBJECTIVES This review underscores the significance of sleep disorders in PD, highlighting their prevalence, impact on disease progression, and the bidirectional relationship between sleep disruption and neurodegeneration. It aims to enhance clinician awareness for better diagnosis and management of sleep-related comorbidities in PD. METHODS A comprehensive literature search was conducted in PubMed and Scopus using key terms such as "sleep disorders", "Parkinson's disease", "REM sleep behavior disorder", "restless legs syndrome", "insomnia", "obstructive sleep apnea", "excessive daytime sleepiness", "circadian rhythm disorders", "sleep and neurodegeneration". RESULTS Sleep disorders are prevalent in PD affecting up to 90% of patients. Conditions such as insomnia, REM sleep behavior disorder, restless legs syndrome, obstructive sleep apnea, excessive daytime sleepiness, and circadian rhythm disorders are commonly reported. These disorders are linked to multifactorial biological mechanisms and are associated with more severe disease phenotypes. Of note, several evidence shows that sleep abnormalities may contribute to neuroinflammation and neurodegeneration, further accelerating the disease course. CONCLUSIONS Sleep disturbances are critical non-motor symptoms in PD. Early diagnosis and tailored management of sleep disorders are essential for improving clinical outcomes and potentially offering neuroprotective benefits.
Collapse
Affiliation(s)
- Elena Antelmi
- Neurology Unit B, AOUIVeronaItaly
- DIMI Department of Engineering and Medicine of InnovationUniversity of VeronaVeronaItaly
| | - Giuseppe Lanza
- Clinical Neurophysiology Research UnitOasi Research Institute‐IRCCSTroinaItaly
- Department of Surgery and Medical‐Surgical SpecialtiesUniversity of CataniaCataniaItaly
| | - Maria Paola Mogavero
- Vita‐Salute San Raffaele UniversityMilanItaly
- San Raffaele Scientific Institute, Division of NeuroscienceSleep Disorders CenterMilanItaly
| | - Gloria Pompea Mingolla
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Giuseppe Plazzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna (ISNB)BolognaItaly
- Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Luigi Ferini‐Strambi
- Vita‐Salute San Raffaele UniversityMilanItaly
- San Raffaele Scientific Institute, Division of NeuroscienceSleep Disorders CenterMilanItaly
| | - Raffaele Ferri
- Clinical Neurophysiology Research UnitOasi Research Institute‐IRCCSTroinaItaly
| | - Michele Tinazzi
- Neurology Unit B, AOUIVeronaItaly
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| |
Collapse
|
12
|
Chudzik A. Machine Learning Recognizes Stages of Parkinson's Disease Using Magnetic Resonance Imaging. SENSORS (BASEL, SWITZERLAND) 2024; 24:8152. [PMID: 39771887 PMCID: PMC11679256 DOI: 10.3390/s24248152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/01/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease (AD) and Parkinson's disease (PD), are debilitating conditions that affect millions worldwide, and the number of cases is expected to rise significantly in the coming years. Because early detection is crucial for effective intervention strategies, this study investigates whether the structural analysis of selected brain regions, including volumes and their spatial relationships obtained from regular T1-weighted MRI scans (N = 168, PPMI database), can model stages of PD using standard machine learning (ML) techniques. Thus, diverse ML models, including Logistic Regression, Random Forest, Support Vector Classifier, and Rough Sets, were trained and evaluated. Models used volumes, Euclidean, and Cosine distances of subcortical brain structures relative to the thalamus to differentiate among control (HC), prodromal (PR), and PD groups. Based on three separate experiments, the Logistic Regression approach was optimal, providing low feature complexity and strong predictive performance (accuracy: 85%, precision: 88%, recall: 85%) in PD-stage recognition. Using interpretable metrics, such as the volume- and centroid-based spatial distances, models achieved high diagnostic accuracy, presenting a promising framework for early-stage PD identification based on MRI scans.
Collapse
Affiliation(s)
- Artur Chudzik
- Faculty of Computer Science, Polish-Japanese Academy of Information Technology, 86 Koszykowa Street, 02-008 Warsaw, Poland
| |
Collapse
|
13
|
Pang MZ, Li HX, Dai XQ, Wang XB, Liu JY, Shen Y, Xu X, Zhong ZM, Wang H, Liu CF, Wang F. Melatonin Ameliorates Abnormal Sleep-Wake Behavior via Facilitating Lipid Metabolism in a Zebrafish Model of Parkinson's Disease. Neurosci Bull 2024; 40:1901-1914. [PMID: 39283564 PMCID: PMC11625096 DOI: 10.1007/s12264-024-01299-8] [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: 12/01/2023] [Accepted: 05/15/2024] [Indexed: 12/08/2024] Open
Abstract
Sleep-wake disorder is one of the most common nonmotor symptoms of Parkinson's disease (PD). Melatonin has the potential to improve sleep-wake disorder, but its mechanism of action is still unclear. Our data showed that melatonin only improved the motor and sleep-wake behavior of a zebrafish PD model when melatonin receptor 1 was present. Thus, we explored the underlying mechanisms by applying a rotenone model. After the PD zebrafish model was induced by 10 nmol/L rotenone, the motor and sleep-wake behavior were assessed. In situ hybridization and real-time quantitative PCR were used to detect the expression of melatonin receptors and lipid-metabolism-related genes. In the PD model, we found abnormal lipid metabolism, which was reversed by melatonin. This may be one of the main pathways for improving PD sleep-wake disorder.
Collapse
Affiliation(s)
- Meng-Zhu Pang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institutes of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Han-Xing Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xue-Qin Dai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institutes of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Bo Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institutes of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun-Yi Liu
- Department of Neurology, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yun Shen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xing Xu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhao-Min Zhong
- Center for Circadian Clocks, Soochow University, Suzhou, 215123, China
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, 215123, China
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institutes of Neuroscience, Soochow University, Suzhou, 215123, China.
- Department of Neurology, Xiongan Xuanwu Hospital, Xiongan, 071700, China.
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institutes of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
14
|
Georgiopoulos C, Werlin A, Lasic S, Hall S, van Westen D, Spotorno N, Hansson O, Nilsson M. Diffusion tensor imaging along the perivascular space: the bias from crossing fibres. Brain Commun 2024; 6:fcae421. [PMID: 39713238 PMCID: PMC11660947 DOI: 10.1093/braincomms/fcae421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/27/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
Non-invasive evaluation of glymphatic function has emerged as a crucial goal in neuroimaging, and diffusion tensor imaging along the perivascular space (DTI-ALPS) has emerged as a candidate method for this purpose. Reduced ALPS index has been suggested to indicate impaired glymphatic function. However, the potential impact of crossing fibres on the ALPS index has not been assessed, which was the aim of this cross-sectional study. For this purpose, we used DTI-ALPS in a cohort with three groups: Parkinson's disease (PD) (n = 60, mean age 63.3 ± 1.5, 33 males), progressive supranuclear palsy (PSP) (n = 17, mean age 70.9 ± 1.5, 9 males) and healthy controls (n = 41, mean age 64.5 ± 8.4, 15 males). The ALPS index was calculated blinded to diagnosis, by manually placing two sets of regions of interest (ROI) on the projection and association fibres of each hemisphere. Annotation was performed twice: once on conventional diffusion-encoded colour maps weighted by fractional anisotropy and once on maps with weights adjusted for high incidence of crossing fibres. PSP patients had significantly lower conventional ALPS indices compared with both healthy controls (right hemisphere: P = 0.009; left hemisphere: P < 0.001) and PD patients (right hemisphere: P = 0.024; left hemisphere: P < 0.001). There were no differences between healthy controls and PD patients. After adjusting the ROI to avoid regions of crossing fibres, the ALPS index significantly decreased in healthy controls (right hemisphere: P < 0.001; left hemisphere: P < 0.001) and PD (right hemisphere: P < 0.001; left hemisphere: P < 0.001). In PSP, the adjusted ALPS index was lower compared with the conventional one only in the right hemisphere (P = 0.047). Overall, this adjustment led to less significant differences among diagnostic groups. Specifically, with the adjusted ALPS index, PSP patients showed significantly lower ALPS index compared with healthy controls (right hemisphere: P = 0.044; left hemisphere: P = 0.029) and PD patients (P = 0.003 for the left hemisphere only). Our results suggest that crossing fibres significantly inflate the ALPS index and should be considered a critical pitfall of this method. This factor could partly explain the variability observed in previous studies. Unlike previous research, we observed no differences between PD and healthy controls, likely because most patients in our cohort were in the early phase of the disease. Thus, the ALPS index may not be a sensitive indicator of glymphatic function at least in the initial stages of neurodegeneration in PD.
Collapse
Affiliation(s)
- Charalampos Georgiopoulos
- Department of Clinical Sciences, Diagnostic Radiology, Medical Faculty, Lund University, 221 85 Lund, Sweden
- Image and Function, Skåne University Hospital, 221 85 Lund, Sweden
| | - Alice Werlin
- Department of Clinical Sciences, Diagnostic Radiology, Medical Faculty, Lund University, 221 85 Lund, Sweden
| | - Samo Lasic
- Department of Clinical Sciences, Diagnostic Radiology, Medical Faculty, Lund University, 221 85 Lund, Sweden
| | - Sara Hall
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Faculty of Medicine, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Danielle van Westen
- Department of Clinical Sciences, Diagnostic Radiology, Medical Faculty, Lund University, 221 85 Lund, Sweden
- Image and Function, Skåne University Hospital, 221 85 Lund, Sweden
| | - Nicola Spotorno
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Faculty of Medicine, Lund University, 211 46 Malmö, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Faculty of Medicine, Lund University, 211 46 Malmö, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Markus Nilsson
- Department of Clinical Sciences, Diagnostic Radiology, Medical Faculty, Lund University, 221 85 Lund, Sweden
| |
Collapse
|
15
|
Khalid Iqbal M, Khan B, Hifsa, YuXuan G, Mujahid M, Kiyani MM, Khan H, Bashir S. The Impact of the Blood-Brain Barrier and Its Dysfunction in Parkinson's Disease: Contributions to Pathogenesis and Progression. ACS OMEGA 2024; 9:45663-45672. [PMID: 39583664 PMCID: PMC11579724 DOI: 10.1021/acsomega.4c06546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
Parkinson's disease (PD) is a brain disorder in which neuronal cells responsible for the release of dopamine, a neurotransmitter that controls movement, are degenerated or impaired in the substantia nigra and basal ganglia. The disease typically affects people over the age of 5 and presents with a variety of motor and nonmotor dysfunctions, which are unique to each person. The impairment of the blood-brain barrier (BBB) and blood retinal barrier (BRB) due to age-related causes such as weakness of tight junctions or rare genetic factors allows several metabolic intermediates to reach and accumulate inside neurons such as Lewy bodies and α-synuclein, disrupting neuronal homeostasis and leading to genetic and epigenetic changes, e.g., damage to the DNA repair system. This perspective highlights the importance of blood barriers, such as the BBB and BRB, in the progression of PD, as the aggregation of Lewy bodies and α-synuclein disrupts neuronal homeostasis. Genetic and epigenetic factors, neuroinflammation, oxidative stress, and mitochondrial dysfunction play crucial roles in the progression of the disease. The implications of these findings are significant; identifying synaptic dysfunction could lead to earlier diagnosis and treatment, while developing targeted therapies focused on preserving synaptic function may slow or halt disease progression. Understanding the various genetic forms of PD could enable more personalized medicine approaches, and using patient-derived midbrain neurons for research may improve the accuracy of PD models due to the implications of an impaired BBB.
Collapse
Affiliation(s)
- Muhammad Khalid Iqbal
- Institute
of Brain Disorders, Department of Physiology, Dalian Medical University, Dalian, Liaoning Province 116044, China
| | - Bakhtawar Khan
- Institute
of Brain Disorders, Department of Physiology, Dalian Medical University, Dalian, Liaoning Province 116044, China
| | - Hifsa
- Department
of Biochemistry, Government College University, Faisalabad 38000, Pakistan
| | - Ge YuXuan
- Institute
of Brain Disorders, Department of Physiology, Dalian Medical University, Dalian, Liaoning Province 116044, China
| | - Muhammad Mujahid
- Department
of Biochemistry, Government College University, Faisalabad 38000, Pakistan
| | - Mubin Mustafa Kiyani
- Shifa
College of Medical Technology, Shifa Tameer-e-Millat
University, Islamabad 44000, Pakistan
| | - Hamid Khan
- Molecular
Biology and Bio Interfaces Engineering Lab, Department of Biological
Sciences, Faculty of Sciences, International
Islamic University Islamabad. H10, Islamabad 44000, Pakistan
| | - Shahid Bashir
- Neuroscience
Center, King Fahad Specialist Hospital Dammam, Dammam 32253, Saudi Arabia
| |
Collapse
|
16
|
Leguizamon M, McKnight CD, Ponzo T, Elenberger J, Eisma JJ, Song AK, Trujillo P, Considine CM, Donahue MJ, Claassen DO, Hett K. Intravenous arachnoid granulation hypertrophy in patients with Parkinson disease. NPJ Parkinsons Dis 2024; 10:177. [PMID: 39304673 DOI: 10.1038/s41531-024-00796-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Intravenous arachnoid granulations (AGs) are protrusions of the arachnoid membrane into the venous lumen and function as contributors to the cerebrospinal fluid (CSF) flow circuit. Patients with Parkinson disease (PD) often present with accumulation of alpha synuclein. Previous works have provided evidence for neurofluid circulation dysfunction in neurodegenerative diseases associated with changes in CSF egress, which may have implications regarding AG morphology. The present study aims to investigate group differences in AG volumetrics between healthy and PD participants, as well as relationships between AG characteristics and clinical assessments. Generalized linear models revealed significant increases in AG volumetrics and number in PD compared to healthy controls. Partial Spearman-rank correlation analyses demonstrated significant relationships between AG metrics and motor and cognitive assessments. Finally, AG volumetrics were positively correlated with objective actigraphy measures of sleep dysfunction, but not self-report sleep symptoms.
Collapse
Affiliation(s)
| | - Colin D McKnight
- Vanderbilt Medical Center, Department of Radiology and Radiological Sciences, Nashville, TN, USA
| | - Tristan Ponzo
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | - Jason Elenberger
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | - Jarrod J Eisma
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | - Alexander K Song
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | - Paula Trujillo
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | | | - Manus J Donahue
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
- Vanderbilt Medical Center, Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, TN, USA
| | - Daniel O Claassen
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA
| | - Kilian Hett
- Vanderbilt Medical Center, Department of Neurology, Nashville, TN, USA.
| |
Collapse
|
17
|
Zhou Y, Liu X, Xu B. Research Progress on the Relationship between Parkinson's Disease and REM Sleep Behavior Disorder. J Integr Neurosci 2024; 23:166. [PMID: 39344226 DOI: 10.31083/j.jin2309166] [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: 01/09/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 10/01/2024] Open
Abstract
An individual's quality of life is greatly affected by Parkinson's disease (PD), a prevalent neurological degenerative condition. Rapid eye movement (REM) sleep behavior disorder (RBD) is a prominent non-motor symptom commonly associated with PD. Previous studies have shown a close relationship between PD and RBD. In addition to being a prodromal symptom of PD, RBD has a major negative impact on the prognosis of PD patients. This intrinsic connection indicates that there is a bidirectional relationship between PD and RBD. This paper provides a comprehensive review of the pathological mechanism related to PD and RBD, including the α-synuclein pathological deposition, abnormal iron metabolism, neuroinflammation, glymphatic system dysfunction and dysbiosis of the gut microbiota. Increasing evidence has shown that RBD patients have the same pathogenic mechanisms that underlie PD, but relatively little research has been done on how RBD contributes to PD progression. Therefore, a more thorough investigation is warranted to characterise how RBD affects the course of PD, in order to prepare for future therapeutic trials.
Collapse
Affiliation(s)
- Yu Zhou
- The Second Clinical Medical College of Zhejiang Chinese Medical University, 310000 Hangzhou, Zhejiang, China
| | - Xiaoli Liu
- Department of Neurology, Zhejiang Hospital Affiliated to Zhejiang University, 310000 Hangzhou, Zhejiang, China
| | - Bin Xu
- Department of Neurology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 310000 Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Liu X, Barisano G, Shao X, Jann K, Ringman JM, Lu H, Arfanakis K, Caprihan A, DeCarli C, Gold BT, Maillard P, Satizabal CL, Fadaee E, Habes M, Stables L, Singh H, Fischl B, van der Kouwe A, Schwab K, Helmer KG, Greenberg SM, Wang DJ. Cross-Vendor Test-Retest Validation of Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) for Evaluating Glymphatic System Function. Aging Dis 2024; 15:1885-1898. [PMID: 37307817 PMCID: PMC11272201 DOI: 10.14336/ad.2023.0321-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/21/2023] [Indexed: 06/14/2023] Open
Abstract
The diffusion tensor image analysis along the perivascular space (DTI-ALPS) method was proposed to evaluate glymphatic system (GS) function. However, few studies have validated its reliability and reproducibility. Fifty participants' DTI data from the MarkVCID consortium were included in this study. Two pipelines by using DSI studio and FSL software were developed for data processing and ALPS index calculation. The ALPS index was obtained by the average of bilateral ALPS index and was used for testing the cross-vendor, inter-rater and test-retest reliability by using R studio software. The ALPS index demonstrated favorable inter-scanner reproducibility (ICC=0.77 to 0.95, P< 0.001), inter-rater reliability (ICC=0.96 to 1, P< 0.001) and test-retest repeatability (ICC=0.89 to 0.95, P< 0.001), offering a potential biomarker for in vivo evaluation of GS function.
Collapse
Affiliation(s)
- Xiaodan Liu
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - Kay Jann
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University, Chicago, IL, USA.
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA.
| | | | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Claudia L Satizabal
- Population Health Sciences and Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Elyas Fadaee
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Lara Stables
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Bruce Fischl
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Computer Science and AI Lab, Cambridge, Massachusetts, USA.
| | - Andre van der Kouwe
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Karl G Helmer
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Danny J.J Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | | |
Collapse
|
19
|
Ryman SG, Vakhtin AA, Mayer AR, van der Horn HJ, Shaff NA, Nitschke SR, Julio KR, Tarawneh RM, Rosenberg GA, Diaz SV, Pirio Richardson SE, Lin HC. Abnormal Cerebrovascular Activity, Perfusion, and Glymphatic Clearance in Lewy Body Diseases. Mov Disord 2024; 39:1258-1268. [PMID: 38817039 PMCID: PMC11341260 DOI: 10.1002/mds.29867] [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: 03/21/2024] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
Cerebrovascular activity is not only crucial to optimal cerebral perfusion, but also plays an important role in the glymphatic clearance of interstitial waste, including α-synuclein. This highlights a need to evaluate how cerebrovascular activity is altered in Lewy body diseases. This review begins by discussing how vascular risk factors and cardiovascular autonomic dysfunction may serve as upstream or direct influences on cerebrovascular activity. We then discuss how patients with Lewy body disease exhibit reduced and delayed cerebrovascular activity, hypoperfusion, and reductions in measures used to capture cerebrospinal fluid flow, suggestive of a reduced capacity for glymphatic clearance. Given the lack of an existing framework, we propose a model by which these processes may foster α-synuclein aggregation and neuroinflammation. Importantly, this review highlights several avenues for future research that may lead to treatments early in the disease course, prior to neurodegeneration. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sephira G Ryman
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Andrei A Vakhtin
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Andrew R Mayer
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Harm Jan van der Horn
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Nicholas A Shaff
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Stephanie R Nitschke
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Kayla R Julio
- Department of Translational Neuroscience, The Mind Research Network, Albuquerque, New Mexico, USA
| | - Rawan M Tarawneh
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
- Cognitive Neurology Section, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Gary A Rosenberg
- Center for Memory and Aging, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Shanna V Diaz
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Sarah E Pirio Richardson
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| | - Henry C Lin
- Department of Internal Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
- New Mexico VA Health Care System, Albuquerque, New Mexico, USA
| |
Collapse
|
20
|
Dong Z, Du X, Wang L, Zou X, Zuo H, Yan Y, Chen G, Cheng O, Zhang Y. Deep cervical lymph nodes in Parkinson's disease and atypical Parkinson's disease: A potential ultrasound biomarker for differential diagnosis. J Cent Nerv Syst Dis 2024; 16:11795735241259429. [PMID: 39086599 PMCID: PMC11289816 DOI: 10.1177/11795735241259429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Background Parkinson's disease (PD) is a common degenerative disease caused by abnormal accumulation of α-synuclein. The glymphatic pathway is essential for removing macromolecular proteins including α-synuclein from the brain, which flows into deep cervical lymph nodes (DCLNs) through meningeal lymphatics. As a terminal station for the cerebral lymphatic system drainage, DCLNs can be easily assessed clinically. Objectives Although the drainage function of the cerebral lymphatic system is impaired in PD, the correlation between DCLNs and PD remains unknown. Design Single-center retrospective cross-sectional study. Methods The size of the DCLNs were measured using ultrasound. The Movement Disorder Society Sponsored Revision Unified Parkinson's Disease Rating Scale and other scales were used to assess PD motor and non-motor symptoms. Results Compared with the healthy control (HC) and the atypical Parkinson's disease (AP) groups, the size of the second and third DCLNs in the Parkinson's disease (PD) group was significantly smaller (P < .05). The width diameter of the third DCLN (DCLN3(y)) was significantly smaller in the PD group than in the AP group (P = .014). DCLN3(y) combined with a variety of clinical features improved the sensitivity of AP identification (sensitivity = .813). Conclusion DCLNs were able to distinguish HC, PD and AP and were mainly located in Robbins ΙΙA level. PD and AP were associated with different factors that influenced the size of the DCLNs. DCLN3(y) plays an important role in differentiating PD from AP, which, combined with other clinical features, has the ability to distinguish PD from AP; in particular, the sensitivity of AP diagnosis was improved.
Collapse
Affiliation(s)
- Zhaoying Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Du
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoya Zou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Yan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guojun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Bispo DPCF, Lins CCSA, Hawkes KL, Tripp S, Khoo TK. The Positive Effects of Physical Activity on Quality of Life in Parkinson's Disease: A Systematic Review. Geriatrics (Basel) 2024; 9:94. [PMID: 39051258 PMCID: PMC11270410 DOI: 10.3390/geriatrics9040094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Physical activity can have positive effects on motor and non-motor symptoms in Parkinson's disease, but its benefits in terms of quality of life and function are uncertain and vary based on the specific forms of activities and interventions. OBJECTIVE We sought to assess the current evidence on the positive effects of physical activity in people with Parkinson's disease and more specifically in relation to its potential benefits for quality of life. METHODS This systematic review was conducted between January and April 2024 via the PubMed, Medline, and Scopus databases. Predetermined search criteria were used that included the following terms: "Parkinson's disease", "quality of life" and "physical activity". RESULTS A total of 1669 articles were identified. After utilizing predetermined criteria, a total of fifteen articles met the selection criteria. Statistically significant improvements in quality of life were found in seven studies. Seven studies demonstrated a significant improvement in non-motor symptoms, while nine studies showed an improvement in motor symptoms. CONCLUSIONS Despite heterogeneity in the study designs, interventions and clinical assessments, the articles identified in this review yielded mostly positive results in relation to physical activities. The findings reflect an improvement in motor and non-motor symptoms may translate to a better quality of life in people with Parkinson's disease.
Collapse
Affiliation(s)
- Dharah P. C. F. Bispo
- School of Medicine & Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
- Neuropsychiatry and Behavioural Sciences Department, Health Sciences Centre, Federal University of Pernambuco, Recife 50670-901, PE, Brazil
- Gerontology Department, Health Sciences Centre, Federal University of Pernambuco, Recife 50670-901, PE, Brazil
| | - Carla C. S. A. Lins
- Gerontology Department, Health Sciences Centre, Federal University of Pernambuco, Recife 50670-901, PE, Brazil
- Anatomy Department, Health Sciences Centre, Federal University of Pernambuco, Recife 50670-901, PE, Brazil
| | - Kelly L. Hawkes
- Northern New South Wales Local Health District, Ballina, NSW 2478, Australia
| | - Shae Tripp
- Northern New South Wales Local Health District, Ballina, NSW 2478, Australia
| | - Tien K. Khoo
- School of Medicine & Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
- Northern New South Wales Local Health District, Ballina, NSW 2478, Australia
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
22
|
Hsu JL, Wei YC, Hsiao IT, Lin KJ, Yen TC, Lu CS, Wang HC, Leemans A, Weng YH, Huang KL. Dominance of Tau Burden in Cortical Over Subcortical Regions Mediates Glymphatic Activity and Clinical Severity in PSP. Clin Nucl Med 2024; 49:387-396. [PMID: 38465965 DOI: 10.1097/rlu.0000000000005141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a tauopathy that involves subcortical regions but also extends to cortical areas. The clinical impact of different tau protein sites and their influence on glymphatic dysfunction have not been investigated. PATIENTS AND METHODS Participants (n = 55; 65.6 ± 7.1 years; 29 women) with PSP (n = 32) and age-matched normal controls (NCs; n = 23) underwent 18 F-Florzolotau tau PET, MRI, PSP Rating Scale (PSPRS), and Mini-Mental State Examination. Cerebellar gray matter (GM) and parametric estimation of reference signal intensity were used as references for tau burden measured by SUV ratios. Glymphatic activity was measured by diffusion tensor image analysis along the perivascular space (DTI-ALPS). RESULTS Parametric estimation of reference signal intensity is a better reference than cerebellar GM to distinguish tau burden between PSP and NCs. PSP patients showed higher cortical and subcortical tau SUV ratios than NCs ( P < 0.001 and <0.001). Cortical and subcortical tau deposition correlated with PSPRS, UPDRS, and Mini-Mental State Examination scores (all P 's < 0.05). Cortical tau deposition was further associated with the DTI-ALPS index and frontal-temporal-parietal GM atrophy. The DTI-ALPS indexes showed a significantly negative correlation with the PSPRS total scores ( P < 0.01). Finally, parietal and occipital lobe tau depositions showed mediating effects between the DTI-ALPS index and PSPRS score. CONCLUSIONS Cortical tau deposition is associated with glymphatic dysfunction and plays a role in mediating glymphatic dysfunction and clinical severity. Our results provide a possible explanation for the worsening of clinical severity in patients with PSP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alexander Leemans
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | |
Collapse
|
23
|
Wen Q, Wang H, Haacke EM, Jiang Q, Hu J. Contribution of Direct Cerebral Vascular Transport in Brain Substance Clearance. Aging Dis 2024; 15:584-600. [PMID: 37611901 PMCID: PMC10917538 DOI: 10.14336/ad.2023.0426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/26/2023] [Indexed: 08/25/2023] Open
Abstract
The accumulation of harmful substances has long been recognized as a likely cause of many neurodegenerative diseases. The two classic brain clearance pathways are cerebrospinal fluid (CSF) and vascular circulation systems. Since the discovery of the glymphatic system, research on the CSF pathway has gained momentum, and impaired CSF clearance has been implicated in virtually all neurodegenerative animal models. However, the contribution of the direct participation of vascular transport across the blood-brain barrier in clearing substances is often ignored in glymphatic papers. Supportive evidence for the direct involvement of parenchymal vasculature in substance clearance is accumulated. First, multiple mechanisms have been proposed for the vascular drainage of exogenous and endogenous substances across the blood-brain barriers. Second, the "traditional" role of arachnoid villi and granulations as the main site for CSF draining into the vasculature system has been questioned. Third, MRI studies using different CSF tracers indicate that parenchymal vasculature directly participates in tracer efflux, consistent with immunohistochemical findings. Here we will review evidence in the literature that supports the direct participation of the parenchymal vascular system in substance clearance, in addition to the CSF clearance pathways.
Collapse
Affiliation(s)
- Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA.
| | - Haoyu Wang
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - E. Mark Haacke
- Department of Radiology, Wayne State University, Detroit, MI 48201 USA.
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202 USA.
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48201 USA.
| |
Collapse
|
24
|
Çavdar S, Altınöz D, Dilan Demir T, Ali Gürses İ, Özcan G. Extracranial transport of brain lymphatics via cranial nerve in human. Neurosci Lett 2024; 827:137737. [PMID: 38519013 DOI: 10.1016/j.neulet.2024.137737] [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: 01/26/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Extracranial waste transport from the brain interstitial fluid to the deep cervical lymph node (dCLN) is not extensively understood. The present study aims to show the cranial nerves that have a role in the transport of brain lymphatics vessels (LVs), their localization, diameter, and number using podoplanin (PDPN) and CD31 immunohistochemistry (IHC) and Western blotting. Cranial nerve samples from 6 human cases (3 cadavers, and 3 autopsies) were evaluated for IHC and 3 autopsies for Western blotting. The IHC staining showed LVs along the optic, olfactory, oculomotor, trigeminal, facial, glossopharyngeal, accessory, and vagus nerves. However, no LVs present along the trochlear, abducens, vestibulocochlear, and hypoglossal nerves. The LVs were predominantly localized at the endoneurium of the cranial nerve that has motor components, and LVs in the cranial nerves that had sensory components were present in all 3 layers. The number of LVs accompanying the olfactory, optic, and trigeminal nerves was classified as numerous; oculomotor, glossopharyngeal, vagus, and accessory was moderate; and facial nerves was few. The largest diameter of LVs was in the epineurium and the smallest one was in the endoneurium. The majority of Western blotting results correlated with the IHC. The present findings suggest that specific cranial nerves with variable quantities provide a pathway for the transport of wastes from the brain to dCLN. Thus, the knowledge of the transport of brain lymphatics along cranial nerves may help understand the pathophysiology of various neurological diseases.
Collapse
Affiliation(s)
- Safiye Çavdar
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey.
| | - Damlasu Altınöz
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Rumelifener Yolu, Istanbul, Turkey
| | - İlke Ali Gürses
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Gülnihal Özcan
- Koç University Research Center for Translational Medicine (KUTTAM), Rumelifener Yolu, Istanbul, Turkey; Department of Medical Pharmacology, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| |
Collapse
|
25
|
Manoutcharian K, Gevorkian G. Recombinant Antibody Fragments for Immunotherapy of Parkinson's Disease. BioDrugs 2024; 38:249-257. [PMID: 38280078 PMCID: PMC10912140 DOI: 10.1007/s40259-024-00646-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2024] [Indexed: 01/29/2024]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder. Multiple genetic and environmental factors leading to progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SN) and consequent depletion of dopamine were described. Current clinical approaches, such as dopamine replacement or deep brain stimulation using surgically implanted probes, provide symptomatic relief but cannot modify disease progression. Therefore, disease-modifying therapeutic tools are urgently needed. Immunotherapy approaches, including passive transfer of protective antibodies and their fragments, have shown therapeutic efficacy in several animal models of neurodegenerative diseases, including PD. Recombinant antibody fragments are promising alternatives to conventional full-length antibodies. Modern computational approaches and molecular biology tools, directed evolution methodology, and the design of tissue-penetrating fusion peptides allowed for the development of recombinant antibody fragments with superior specificity and affinity, reduced immunogenicity, the capacity to target hidden epitopes and cross the blood-brain barrier (BBB), higher solubility and stability, the ability to refold after heat denaturation, and inexpensive large-scale production. In addition, antibody fragments do not induce microglia Fcγ receptor (FcγR)-mediated proinflammatory response and tissue damage in the central nervous system (CNS), because they lack the Fc portion of the immunoglobulin molecule. In the present review, we summarized data on recombinant antibody fragments evaluated as immunotherapeutics in preclinical models of PD and discussed their potential for developing therapeutic and preventive protocols for patients with PD.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, CP 04510, Mexico, DF, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, CP 04510, Mexico, DF, Mexico.
| |
Collapse
|
26
|
Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Elewa YHA, AL‐Farga A, Aqlan F, Zahran MH, Batiha GE. Sleep disorders cause Parkinson's disease or the reverse is true: Good GABA good night. CNS Neurosci Ther 2024; 30:e14521. [PMID: 38491789 PMCID: PMC10943276 DOI: 10.1111/cns.14521] [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: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative brain disease due to degeneration of dopaminergic neurons (DNs) presented with motor and non-motor symptoms. PD symptoms are developed in response to the disturbance of diverse neurotransmitters including γ-aminobutyric acid (GABA). GABA has a neuroprotective effect against PD neuropathology by protecting DNs in the substantia nigra pars compacta (SNpc). It has been shown that the degeneration of GABAergic neurons is linked with the degeneration of DNs and the progression of motor and non-motor PD symptoms. GABA neurotransmission is a necessary pathway for normal sleep patterns, thus deregulation of GABAergic neurotransmission in PD could be the potential cause of sleep disorders in PD. AIM Sleep disorders affect GABA neurotransmission leading to memory and cognitive dysfunction in PD. For example, insomnia and short sleep duration are associated with a reduction of brain GABA levels. Moreover, PD-related disorders including rigidity and nocturia influence sleep patterns leading to fragmented sleep which may also affect PD neuropathology. However, the mechanistic role of GABA in PD neuropathology regarding motor and non-motor symptoms is not fully elucidated. Therefore, this narrative review aims to clarify the mechanistic role of GABA in PD neuropathology mainly in sleep disorders, and how good GABA improves PD. In addition, this review of published articles tries to elucidate how sleep disorders such as insomnia and REM sleep behavior disorder (RBD) affect PD neuropathology and severity. The present review has many limitations including the paucity of prospective studies and most findings are taken from observational and preclinical studies. GABA involvement in the pathogenesis of PD has been recently discussed by recent studies. Therefore, future prospective studies regarding the use of GABA agonists in the management of PD are suggested to observe their distinct effects on motor and non-motor symptoms. CONCLUSION There is a bidirectional relationship between the pathogenesis of PD and sleep disorders which might be due to GABA deregulation.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ammar AL‐Farga
- Biochemistry Department, College of SciencesUniversity of JeddahJeddahSaudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of SciencesIbb UniversityIbb GovernorateYemen
| | | | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurEgypt
| |
Collapse
|
27
|
Lapshina KV, Ekimova IV. Aquaporin-4 and Parkinson's Disease. Int J Mol Sci 2024; 25:1672. [PMID: 38338949 PMCID: PMC10855351 DOI: 10.3390/ijms25031672] [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: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The water-selective channel aquaporin-4 (AQP4) is implicated in water homeostasis and the functioning of the glymphatic system, which eliminates various metabolites from the brain tissue, including amyloidogenic proteins. Misfolding of the α-synuclein protein and its post-translational modifications play a crucial role in the development of Parkinson's disease (PD) and other synucleopathies, leading to the formation of cytotoxic oligomers and aggregates that cause neurodegeneration. Human and animal studies have shown an interconnection between AQP4 dysfunction and α-synuclein accumulation; however, the specific role of AQP4 in these mechanisms remains unclear. This review summarizes the current knowledge on the role of AQP4 dysfunction in the progression of α-synuclein pathology, considering the possible effects of AQP4 dysregulation on brain molecular mechanisms that can impact α-synuclein modification, accumulation and aggregation. It also highlights future directions that can help study the role of AQP4 in the functioning of the protective mechanisms of the brain during the development of PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ksenia V. Lapshina
- Laboratory of Comparative Thermophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 194223 Saint Petersburg, Russia;
| | | |
Collapse
|
28
|
Manoutcharian K, Gevorkian G. Recombinant Antibody Fragments for Neurological Disorders: An Update. Curr Neuropharmacol 2024; 22:2157-2167. [PMID: 37646225 PMCID: PMC11337690 DOI: 10.2174/1570159x21666230830142554] [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: 04/11/2023] [Revised: 06/15/2023] [Accepted: 07/13/2023] [Indexed: 09/01/2023] Open
Abstract
Recombinant antibody fragments are promising alternatives to full-length immunoglobulins, creating big opportunities for the pharmaceutical industry. Nowadays, antibody fragments such as antigen-binding fragments (Fab), single-chain fragment variable (scFv), single-domain antibodies (sdAbs), and bispecific antibodies (bsAbs) are being evaluated as diagnostics or therapeutics in preclinical models and in clinical trials. Immunotherapy approaches, including passive transfer of protective antibodies, have shown therapeutic efficacy in several animal models of Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), Huntington's disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). There are various antibodies approved by the Food and Drug Administration (FDA) for treating multiple sclerosis and two amyloid beta-specific humanized antibodies, Aducanumab and Lecanemab, for AD. Our previous review summarized data on recombinant antibodies evaluated in pre-clinical models for immunotherapy of neurodegenerative diseases. Here, we explore recent studies in this fascinating research field, give an update on new preventive and therapeutic applications of recombinant antibody fragments for neurological disorders and discuss the potential of antibody fragments for developing novel approaches for crossing the blood-brain barrier (BBB) and targeting cells and molecules of interest in the brain.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Mexico
| |
Collapse
|
29
|
Zhang J, Liu S, Wu Y, Tang Z, Wu Y, Qi Y, Dong F, Wang Y. Enlarged Perivascular Space and Index for Diffusivity Along the Perivascular Space as Emerging Neuroimaging Biomarkers of Neurological Diseases. Cell Mol Neurobiol 2023; 44:14. [PMID: 38158515 PMCID: PMC11407189 DOI: 10.1007/s10571-023-01440-7] [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: 03/06/2023] [Accepted: 11/12/2023] [Indexed: 01/03/2024]
Abstract
The existence of lymphatic vessels or similar clearance systems in the central nervous system (CNS) that transport nutrients and remove cellular waste is a neuroscientific question of great significance. As the brain is the most metabolically active organ in the body, there is likely to be a potential correlation between its clearance system and the pathological state of the CNS. Until recently the successive discoveries of the glymphatic system and the meningeal lymphatics solved this puzzle. This article reviews the basic anatomy and physiology of the glymphatic system. Imaging techniques to visualize the function of the glymphatic system mainly including post-contrast imaging techniques, indirect lymphatic assessment by detecting increased perivascular space, and diffusion tensor image analysis along the perivascular space (DTI-ALPS) are discussed. The pathological link between glymphatic system dysfunction and neurological disorders is the key point, focusing on the enlarged perivascular space (EPVS) and the index of diffusivity along the perivascular space (ALPS index), which may represent the activity of the glymphatic system as possible clinical neuroimaging biomarkers of neurological disorders. The pathological link between glymphatic system dysfunction and neurological disorders is the key point, focusing on the enlarged perivascular space (EPVS) and the index for of diffusivity along the perivascular space (ALPS index), which may represent the activity of the glymphatic system as possible clinical neuroimaging biomarkers of neurological disorders.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaqi Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhijian Tang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yasong Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fangyong Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
30
|
He Y, Guan J, Lai L, Zhang X, Chen B, Wang X, Wu R. Imaging of brain clearance pathways via MRI assessment of the glymphatic system. Aging (Albany NY) 2023; 15:14945-14956. [PMID: 38149988 PMCID: PMC10781494 DOI: 10.18632/aging.205322] [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: 07/25/2023] [Accepted: 11/03/2023] [Indexed: 12/28/2023]
Abstract
Glymphatic clearance dysfunction may play an important role in a variety of neurodegenerative diseases and the progression of ageing. However, in vivo imaging of the glymphatic system is challenging. In this study, we describe an MRI method based on chemical exchange saturation transfer (CEST) of the Angiopep-2 probe to visualize the clearance function of the glymphatic system. We injected rats with Angiopep-2 via the tail vein and performed in vivo MRI at 7 T to track differences in Angiopep-2 signal changes; we then applied the same principles in a bilateral deep cervical lymph node ligation rat model and in ageing rats. We demonstrated the feasibility of Angiopep-2 CEST for visualizing the clearance function of the glymphatic system. Finally, a pathological assessment was performed. Within the model group, the deep cervical lymph node ligation group and the ageing group showed higher CEST signal than the control group. We conclude that this new MRI method can visualize clearance in the glymphatic system.
Collapse
Affiliation(s)
- Yi He
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Department of Ultrasound, Shantou Central Hospital, Shantou, Guangdong, China
| | - Jitian Guan
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Lingfeng Lai
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xiaolei Zhang
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Beibei Chen
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xueqing Wang
- Department of Ultrasound, Shantou Central Hospital, Shantou, Guangdong, China
| | - Renhua Wu
- Department of Medical Imaging, Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
31
|
Kumari N, Anand S, Shah K, Chauhan NS, Sethiya NK, Singhal M. Emerging Role of Plant-Based Bioactive Compounds as Therapeutics in Parkinson's Disease. Molecules 2023; 28:7588. [PMID: 38005310 PMCID: PMC10673433 DOI: 10.3390/molecules28227588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Neurological ailments, including stroke, Alzheimer's disease (AD), epilepsy, Parkinson's disease (PD), and other related diseases, have affected around 1 billion people globally to date. PD stands second among the common neurodegenerative diseases caused as a result of dopaminergic neuron loss in the midbrain's substantia nigra regions. It affects cognitive and motor activities, resulting in tremors during rest, slow movement, and muscle stiffness. There are various traditional approaches for the management of PD, but they provide only symptomatic relief. Thus, a survey for finding new biomolecules or substances exhibiting the therapeutic potential to patients with PD is the main focus of present-day research. Medicinal plants, herbal formulations, and natural bioactive molecules have been gaining much more attention in recent years as synthetic molecules orchestrate a number of undesired effects. Several in vitro, in vivo, and in silico studies in the recent past have demonstrated the therapeutic potential of medicinal plants, herbal formulations, and plant-based bioactives. Among the plant-based bioactives, polyphenols, terpenes, and alkaloids are of particular interest due to their potent anti-inflammatory, antioxidant, and brain-health-promoting properties. Further, there are no concise, elaborated articles comprising updated mechanism-of-action-based reviews of the published literature on potent, recently investigated (2019-2023) medicinal plants, herbal formulations, and plant based-bioactive molecules, including polyphenols, terpenes, and alkaloids, as a method for the management of PD. Therefore, we designed the current review to provide an illustration of the efficacious role of various medicinal plants, herbal formulations, and bioactives (polyphenols, terpenes, and alkaloids) that can become potential therapeutics against PD with greater specificity, target approachability, bioavailability, and safety to the host. This information can be further utilized in the future to develop several value-added formulations and nutraceutical products to achieve the desired safety and efficacy for the management of PD.
Collapse
Affiliation(s)
- Nitu Kumari
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, Karnataka, India;
| | - Santosh Anand
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, Karnataka, India;
| | - Kamal Shah
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, Uttar Pradesh, India;
| | | | - Neeraj K. Sethiya
- Faculty of Pharmacy, School of Pharmaceutical and Populations Health Informatics, DIT University, Dehradun 248009, Uttarakhand, India;
| | - Manmohan Singhal
- Faculty of Pharmacy, School of Pharmaceutical and Populations Health Informatics, DIT University, Dehradun 248009, Uttarakhand, India;
| |
Collapse
|
32
|
Gnarra O, Calvello C, Schirinzi T, Beozzo F, De Masi C, Spanetta M, Fernandes M, Grillo P, Cerroni R, Pierantozzi M, Bassetti CLA, Mercuri NB, Stefani A, Liguori C. Exploring the Association Linking Head Position and Sleep Architecture to Motor Impairment in Parkinson's Disease: An Exploratory Study. J Pers Med 2023; 13:1591. [PMID: 38003906 PMCID: PMC10671918 DOI: 10.3390/jpm13111591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Patients with Parkinson's disease (PD) tend to sleep more frequently in the supine position and less often change head and body position during sleep. Besides sleep quality and continuity, head and body positions are crucial for glymphatic system (GS) activity. This pilot study evaluated sleep architecture and head position during each sleep stage in idiopathic PD patients without cognitive impairment, correlating sleep data to patients' motor and non-motor symptoms (NMS). All patients underwent the multi-night recordings, which were acquired using the Sleep Profiler headband. Sleep parameters, sleep time in each head position, and percentage of slow wave activity (SWA) in sleep, stage 3 of non-REM sleep (N3), and REM sleep in the supine position were extracted. Lastly, correlations with motor impairment and NMS were performed. Twenty PD patients (65.7 ± 8.6 y.o, ten women) were included. Sleep architecture did not change across the different nights of recording and showed the prevalence of sleep performed in the supine position. In addition, SWA and N3 were more frequently in the supine head position, and N3 in the supine decubitus correlated with REM sleep performed in the same position; this latter correlated with the disease duration (correlation coefficient = 0.48, p-value = 0.03) and motor impairment (correlation coefficient = 0.53, p-value = 0.02). These preliminary results demonstrated the importance of monitoring sleep in PD patients, supporting the need for preventive strategies in clinical practice for maintaining the lateral head position during the crucial sleep stages (SWA, N3, REM), essential for permitting the GS function and activity and ensuring brain health.
Collapse
Affiliation(s)
- Oriella Gnarra
- Sleep-Wake-Epilepsy Center, Department of Neurology, University Hospital of Bern, 3010 Bern, Switzerland; (O.G.); (C.L.A.B.)
- Sensory-Motor Systems Lab, Institute of Robotics and Intelligent Systems, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Carmen Calvello
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | - Francesca Beozzo
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
| | - Claudia De Masi
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | | | - Mariana Fernandes
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
| | - Piergiorgio Grillo
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | - Rocco Cerroni
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | - Mariangela Pierantozzi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | - Claudio L. A. Bassetti
- Sleep-Wake-Epilepsy Center, Department of Neurology, University Hospital of Bern, 3010 Bern, Switzerland; (O.G.); (C.L.A.B.)
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
- Neurology Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Alessandro Stefani
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
- Parkinson’s Disease Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy; (C.D.M.); (P.G.); (R.C.)
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.C.); (T.S.); (F.B.); (M.P.); (N.B.M.); (A.S.)
- Neurology Unit, University Hospital of Rome “Tor Vergata”, 00133 Rome, Italy
| |
Collapse
|
33
|
Çavdar S, Köse B, Altınöz D, Söyler G, Cingöz A, Gürses İA, Özkan M, Aslıyüksek H, Çakır H. Lymphatic Vessels Accompanying Dorsal and Basal Dural Sinuses in the Human Brain. J Chem Neuroanat 2023; 134:102357. [PMID: 39492448 DOI: 10.1016/j.jchemneu.2023.102357] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/02/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Recent investigations showed the presence of meningeal lymphatic vessels (mLVs) along the superior sagittal and transverse dural sinuses which drain both fluid and immune cells from the cerebrospinal fluid (CSF) to the deep cervical lymph nodes. This study uses immunohistochemistry (IHC) and the Western Blot technique to show the presence of mLV accompanying the dorsal (superior sagittal, inferior sagittal, transverse, sigmoid, and straight) and basal (cavernous, sphenoparietal, superior, and inferior petrosal) dural sinuses in the human brain. Samples for IHC were obtained from dorsal and basal meningeal dural sinuses of 3 human cadavers and 3 autopsies. Routine histological techniques were carried out for the specimens. Podoplanin (PDPN, lymphatic vessel endothelial cell marker) and CD31 (vascular endothelial cell marker) IHC staining were applied to the 5µm thick paraffin sections. Furthermore, PDPN and CD31 protein expressions were evaluated using Western Blot to the tissue samples from the same regions of 4 autopsies. Two consecutive sections from each sinus were PDPN, and CD31 was stained to differentiate blood vessels (BV) from mLV. The IHC staining showed the presence of mLVs accompanying both dorsal and basal dural sinuses. The mLVs accompanying the dorsal dural sinuses had a larger dimensions range compared to the basal dural sinuses. However, the number of mLVs along the basal dural sinuses was more than the mLVs along the dorsal ones. Further, fluid channels were closely localized to the mLV, with varying diameters and densities. Western Blotting technique showed the presence of PDPN expression in both dorsal and basal dural sinus samples. The knowledge of the presence of mLV along both dorsal and basal dural sinuses in humans can increase the understanding of how mLV contributes to the brain lymphatic circulation and may help understand the neuropathophysiological processes of various neurological diseases.
Collapse
Affiliation(s)
- Safiye Çavdar
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey.
| | - Büşra Köse
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Damlasu Altınöz
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Gizem Söyler
- Department of Histology, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Ahmet Cingöz
- Koç University Research Center for Translational Medicine, Istanbul, Turkey, 34450
| | - İlke Ali Gürses
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Mazhar Özkan
- Department of Anatomy, Tekirdağ Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | | | - Halit Çakır
- The Council of Forensic Medicine (ATK), Istanbul, Turkey
| |
Collapse
|
34
|
de Bergeyck R, Geoffroy PA. Insomnia in neurological disorders: Prevalence, mechanisms, impact and treatment approaches. Rev Neurol (Paris) 2023; 179:767-781. [PMID: 37620177 DOI: 10.1016/j.neurol.2023.08.008] [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: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023]
Abstract
Insomnia is more prevalent in neurological disorders compared to the general population, with rates ranging from 11 to 74.2% in neurodegenerative disorders, 20 to 37% in vascular diseases, 13.3 to 50% in inflammatory diseases, 28.9 to 74.4% in epilepsy, and nearly 70% in migraines. Insomnia in neurological disorders stems from a variety of factors, encompassing physical and neuropsychiatric factors, behavioral patterns, and disruptions in the biological clock and circadian rhythm. There are bidirectional connections between neurological disorders and insomnia. Insomnia in neurological disorders worsens symptoms, resulting in heightened depressive symptoms, elevated mortality rates, reduced quality of life, and intensified acute symptoms. Managing comorbid sleep disorders, especially in the presence of psychiatric comorbidities, is crucial. Cognitive behavioral therapy for insomnia (CBT-I) is the first-line recommendation for insomnia management in neurological disorders. Other treatments are second-line strategies. Melatonin may demonstrate effectiveness in addressing insomnia, with soporific and chronobiotic effects. Furthermore, it has the potential to alleviate "sundowning" and behavioral disturbances, while generally being well-tolerated. Other treatment options that may be of interest include morning bright light therapy, sedative antidepressants, new orexin dual antagonists and levodopa specifically indicated for Parkinson's disease. Benzodiazepines and z-drugs can be used primarily during acute phases to prevent pharmacotolerance and minimize side effects. However, they should be avoided in patients with neurological disorders and not used in patients over 75 years old due to the risk of falls and confusion. In neurological disorders, insomnia has a profound impact on daytime functioning, making its management crucial. Effective treatment can result in improved outcomes, and additional research is necessary to investigate alternative therapeutic options and enhance patient care.
Collapse
Affiliation(s)
- R de Bergeyck
- Centre ChronoS, GHU Paris - Psychiatry Neurosciences, 1, rue Cabanis, 75014 Paris, France.
| | - P A Geoffroy
- Centre ChronoS, GHU Paris - Psychiatry Neurosciences, 1, rue Cabanis, 75014 Paris, France; Département de psychiatrie et d'addictologie, DMU Neurosciences, GHU Paris Nord, hôpital Bichat-Claude-Bernard, AP-HP, 75018 Paris, France; Université Paris Cité, NeuroDiderot, Inserm U1141, 75019 Paris, France; CNRS UPR 3212, Institute for Cellular and Integrative Neurosciences, 5, rue Blaise-Pascal, 67000 Strasbourg, France
| |
Collapse
|
35
|
Luo Y, Liu J, Chen D, Liu M, Yuan Y, Hu J, Wu J, Wang F, Liu C, Chen J, Mao C. How sleep quality affects activities of daily living in Parkinson's disease: the mediating role of disease severity and the moderating role of cognition. Front Aging Neurosci 2023; 15:1238588. [PMID: 37842121 PMCID: PMC10570447 DOI: 10.3389/fnagi.2023.1238588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Objective The aim of this study was to explore the influential mechanism of the relationship between sleep quality and activities of daily living (ADL) in patients with Parkinson's disease (PD), we hypothesized disease severity as a mediator and assumed the mediating process was regulated by cognition. Methods 194 individuals with PD (95 women and 99 men) were enrolled in study. The Pittsburgh Sleep Quality Index (PSQI) was used to assess sleep quality of PD patients. Patients' ADL, disease severity and cognition were measured by the Unified Parkinson's Disease Rating Scale-II (UPDRSII), Hoehn-Yahr (H-Y) Scale, and Mini-Mental State Examination (MMSE). We investigated the mediating role of disease severity and the moderating effect of cognition on the association between sleep quality and ADL in PD patients. Results The score of UPDRSII was positively correlated with the score of PSQI and H-Y stage, while the score of MMSE was negatively correlated with the score of H-Y stage and UPDRSII. Sleep quality predicts disease severity, and disease severity predicts ADL. Disease severity mediated the relationship between sleep quality and ADL, and the mediating effect was 0.179. Cognition alone did not affect ADL, but the interaction between disease severity and cognition was significantly affected ADL, confirming the moderating effect of cognition in PD patients. Conclusion Disease severity mediated the association between sleep quality and ADL, good cognition significantly reduced disease severity's mediating influence on the relationship between sleep quality and ADL. Our study indicated a close relationship between ADL and sleep and cognition in PD, and also provided new insights into the overall management of PD and a better quality of life of PD patients.
Collapse
Affiliation(s)
- Yajun Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junyi Liu
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Dongqin Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Manhua Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Yuan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingzhe Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayu Wu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and 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
| | - Chunfeng Liu
- Department of Neurology and 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
| | - Juping Chen
- Department of Neurology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Chengjie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
He P, Gao Y, Shi L, Li Y, Jiang S, Tie Z, Qiu Y, Ma G, Zhang Y, Nie K, Wang L. Motor progression phenotypes in early-stage Parkinson's Disease: A clinical prediction model and the role of glymphatic system imaging biomarkers. Neurosci Lett 2023; 814:137435. [PMID: 37562710 DOI: 10.1016/j.neulet.2023.137435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Substantial heterogeneity of motor symptoms in Parkinson's disease (PD) poses a challenge to disease prediction. OBJECTIVES The aim of this study was to construct a nomogram model that can distinguish different longitudinal trajectories of motor symptom changes in early-stage PD patients. METHODS Data on 90 patients with 5-years of follow-up were collected from the Parkinson's Progression Marker Initiative (PPMI) cohort. We used a latent class mixed modeling (LCMM) to identify distinct progression patterns of motor symptoms, and backward stepwise logistic regression with baseline information was conducted to identify the potential predictors for motor trajectory and to develop a nomogram. The performance of the nomogram model was then evaluated using the optimism-corrected C-index for internal validation, the area under the curve (AUC) of the receiver operating characteristic (ROC) curve for discrimination, the calibration curve for predictive accuracy, and decision curve analysis (DCA) for its clinical value. RESULTS We identified two trajectories for motor progression patterns. The first, Class 1 (Motor deteriorated group), was characterized by sustained, continuously worsening motor symptoms, and the second, Class 2 (Motor stable group), had stable motor symptoms throughout the follow-up period. The best combination of 7 baseline variables was identified and assembled into the nomogram: Scopa-AUT [odds ratio (OR), 1.11; p = 0.091], Letter number sequencing (LNS) (OR, 0.76; p = 0.068), the asymmetry index of putamen (OR, 0.95; p = 0.034), mean caudate uptake (OR, 0.14; p = 0.086), CSF pTau/α-synuclein (OR, 0.00; p = 0.011), CSF tTau/Aβ (OR, 25434806; p = 0.025), and the index for diffusion tensor image analysis along the perivascular space (ALPS-index) (OR, 0.02; p = 0.030). The nomogram achieved good discrimination, with an original AUC of 0.901 (95% CI, 0.813-0.989), and the bias-corrected concordance index (C-index) with 1,000 bootstraps was 0.834. The calibration curve and DCA also suggested both the high accuracy and clinical usefulness of the nomogram, respectively. CONCLUSIONS This study proposes an effective nomogram to predict different motor progression patterns in early-stage PD. Furthermore, the imaging biomarker indicating glymphatic function could be an independent predictive factor for PD motor progression.
Collapse
Affiliation(s)
- Peikun He
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China; BrainNow Research Institute, Shenzhen, Guangdong Province, China
| | - Yanyi Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shuolin Jiang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zihui Tie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yihui Qiu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guixian Ma
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Kun Nie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Lijuan Wang
- School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
37
|
Buongiorno M, Marzal C, Fernandez M, Cullell N, de Mena L, Sánchez-Benavides G, de la Sierra A, Krupinski J, Compta Y. Altered sleep and neurovascular dysfunction in alpha-synucleinopathies: the perfect storm for glymphatic failure. Front Aging Neurosci 2023; 15:1251755. [PMID: 37693650 PMCID: PMC10484002 DOI: 10.3389/fnagi.2023.1251755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Clinical and cognitive progression in alpha-synucleinopathies is highly heterogeneous. While some patients remain stable over long periods of time, other suffer early dementia or fast motor deterioration. Sleep disturbances and nocturnal blood pressure abnormalities have been identified as independent risk factors for clinical progression but a mechanistic explanation linking both aspects is lacking. We hypothesize that impaired glymphatic system might play a key role on clinical progression. Glymphatic system clears brain waste during specific sleep stages, being blood pressure the motive force that propels the interstitial fluid through brain tissue to remove protein waste. Thus, the combination of severe sleep alterations, such as REM sleep behavioral disorder, and lack of the physiological nocturnal decrease of blood pressure due to severe dysautonomia may constitute the perfect storm for glymphatic failure, causing increased abnormal protein aggregation and spreading. In Lewy body disorders (Parkinson's disease and dementia with Lewy bodies) the increment of intraneuronal alpha-synuclein and extracellular amyloid-β would lead to cognitive deterioration, while in multisystemic atrophy, increased pathology in oligodendroglia would relate to the faster and malignant motor progression. We present a research model that may help in developing studies aiming to elucidate the role of glymphatic function and associated factors mainly in alpha-synucleinopathies, but that could be relevant also for other protein accumulation-related neurodegenerative diseases. If the model is proven to be useful could open new lines for treatments targeting glymphatic function (for example through control of nocturnal blood pressure) with the objective to ameliorate cognitive and motor progression in alpha-synucleinopathies.
Collapse
Affiliation(s)
- Mariateresa Buongiorno
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Clara Marzal
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Manel Fernandez
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Institut de Neurociències (UBNeuro), Universitat de Barcelona, Barcelona, Spain
| | - Natalia Cullell
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Lorena de Mena
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Institut de Neurociències (UBNeuro), Universitat de Barcelona, Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Alejandro de la Sierra
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Jerzy Krupinski
- Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MútuaTerrassa, Terrassa, Spain
- Department of Life Sciences John Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Yaroslau Compta
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, CIBERNED (CB06/05/0018-ISCIII), ERN-RND, UBNeuro Institut Clínic de Neurociències (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Cochen De Cock V, Dodet P, Leu‐Semenescu S, Aerts C, Abril B, Castelnovo G, Landragin N, Drapier S, Olivet H, Corbillé A, Leclair‐Visonneau L, Anheim M, Vidailhet M, Arnulf I, Doulazmi M, Roze E. Night-Time Apomorphine Infusion: Who Are the Best Candidates? Mov Disord Clin Pract 2023; 10:1192-1197. [PMID: 37635769 PMCID: PMC10450238 DOI: 10.1002/mdc3.13799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 08/29/2023] Open
Abstract
Background We recently demonstrated in a randomized controlled trial (APOMORPHEE, NCT02940912) that night-time only subcutaneous apomorphine infusion improves sleep disturbances and insomnia in patients with advanced Parkinson's disease and moderate to severe insomnia. Objectives To identify the best candidates for receiving night-time only subcutaneous apomorphine infusion in routine care. Methods In this post-hoc analysis of APOMORPHEE, we compared the characteristics of patients according to whether they chose to continue night-time only subcutaneous apomorphine infusion at the end of the study period or not. Results Half of the patients (22/42) chose to continue the treatment. Off duration (day or night), painful Off dystonia, and insomnia severity at baseline were associated with night-time only apomorphine continuation. Multivariate analysis retained only Off duration as an independent predictor of continuation. Conclusions The best candidates for night-time only apomorphine are patients with severe and prolonged Off periods (day or night) and severe insomnia.
Collapse
Affiliation(s)
- Valérie Cochen De Cock
- Sleep and Neurology DepartmentBeau Soleil ClinicMontpellierFrance
- EuroMov Digital Health in MotionUniv Montpellier, IMT Mines AlèsMontpellierFrance
| | - Pauline Dodet
- Sleep Disorders UnitPitié‐Salpêtrière University HospitalParisFrance
| | | | - Cécile Aerts
- Sleep and Neurology DepartmentBeau Soleil ClinicMontpellierFrance
- EuroMov Digital Health in MotionUniv Montpellier, IMT Mines AlèsMontpellierFrance
| | - Beatriz Abril
- Sleep DepartmentUniversity Hospital of NîmesNîmesFrance
| | | | | | - Sophie Drapier
- Department of NeurologyPontchaillou INSERM CIC1414RennesFrance
| | - Hélène Olivet
- Sleep DepartmentPolyclinique Rennes Saint‐LaurentRennesFrance
| | | | - Laurène Leclair‐Visonneau
- Clinical Neurophysiology DepartmentUniversity Hospital of Nantes, INSERM U1235, Nantes UniversityNantesFrance
| | - Mathieu Anheim
- Department of NeurologyUniversity Hospital of StrasbourgStrasbourgFrance
- Genetic Institute and Molecular and Cellular Biology (IGBMC), INSERM‐U964/CNRS‐UMR7104/ Strasbourg UniversityIllkirchFrance
| | - Marie Vidailhet
- AP‐HP, Salpetriere Hospital, DMU Neuroscience 6ParisFrance
- Sorbonne University, Paris Brain Institute, Inserm, CNRSParisFrance
| | - Isabelle Arnulf
- Sleep Disorders UnitPitié‐Salpêtrière University HospitalParisFrance
- Sorbonne University, Paris Brain Institute, Inserm, CNRSParisFrance
| | - Mohamed Doulazmi
- Adaptation Biologique et Vieillissement (UMR8256), Institut de Biologie Paris Seine, Sorbonne University, CNRSParisFrance
| | - Emmanuel Roze
- AP‐HP, Salpetriere Hospital, DMU Neuroscience 6ParisFrance
- Sorbonne University, Paris Brain Institute, Inserm, CNRSParisFrance
| | | |
Collapse
|
39
|
Alrouji M, Al-Kuraishy HM, Al-Gareeb AI, Zaafar D, Batiha GES. Orexin pathway in Parkinson's disease: a review. Mol Biol Rep 2023; 50:6107-6120. [PMID: 37155018 DOI: 10.1007/s11033-023-08459-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease (NDD) caused by dopaminergic neuron degeneration in the substantia nigra (SN). Orexin is a neuropeptide that plays a role in the pathogenesis of PD. Orexin has neuroprotective properties in dopaminergic neurons. In PD neuropathology, there is also degeneration of orexinergic neurons in the hypothalamus, in addition to dopaminergic neurons. However, the loss of orexinergic neurons in PD began after the degeneration of dopaminergic neurons. Reduced activity of orexinergic neurons has been linked to developing and progressing motor and non-motor symptoms in PD. In addition, the dysregulation of the orexin pathway is linked to the development of sleep disorders. The hypothalamic orexin pathway regulates various aspects of PD neuropathology at the cellular, subcellular, and molecular levels. Finally, non-motor symptoms, particularly insomnia and disturbed sleep, promote neuroinflammation and the accumulation of neurotoxic proteins as a result of defects in autophagy, endoplasmic reticulum (ER) stress, and the glymphatic system. As a result, this review aimed to highlight the potential role of orexin in PD neuropathology.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Al Beheira, Egypt.
| |
Collapse
|
40
|
Que M, Li Y, Wang X, Zhan G, Luo X, Zhou Z. Role of astrocytes in sleep deprivation: accomplices, resisters, or bystanders? Front Cell Neurosci 2023; 17:1188306. [PMID: 37435045 PMCID: PMC10330732 DOI: 10.3389/fncel.2023.1188306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Sleep plays an essential role in all studied animals with a nervous system. However, sleep deprivation leads to various pathological changes and neurobehavioral problems. Astrocytes are the most abundant cells in the brain and are involved in various important functions, including neurotransmitter and ion homeostasis, synaptic and neuronal modulation, and blood-brain barrier maintenance; furthermore, they are associated with numerous neurodegenerative diseases, pain, and mood disorders. Moreover, astrocytes are increasingly being recognized as vital contributors to the regulation of sleep-wake cycles, both locally and in specific neural circuits. In this review, we begin by describing the role of astrocytes in regulating sleep and circadian rhythms, focusing on: (i) neuronal activity; (ii) metabolism; (iii) the glymphatic system; (iv) neuroinflammation; and (v) astrocyte-microglia cross-talk. Moreover, we review the role of astrocytes in sleep deprivation comorbidities and sleep deprivation-related brain disorders. Finally, we discuss potential interventions targeting astrocytes to prevent or treat sleep deprivation-related brain disorders. Pursuing these questions would pave the way for a deeper understanding of the cellular and neural mechanisms underlying sleep deprivation-comorbid brain disorders.
Collapse
Affiliation(s)
- Mengxin Que
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Zhou
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Pamphlett R, Bishop DP. The toxic metal hypothesis for neurological disorders. Front Neurol 2023; 14:1173779. [PMID: 37426441 PMCID: PMC10328356 DOI: 10.3389/fneur.2023.1173779] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/30/2023] [Indexed: 07/11/2023] Open
Abstract
Multiple sclerosis and the major sporadic neurogenerative disorders, amyotrophic lateral sclerosis, Parkinson disease, and Alzheimer disease are considered to have both genetic and environmental components. Advances have been made in finding genetic predispositions to these disorders, but it has been difficult to pin down environmental agents that trigger them. Environmental toxic metals have been implicated in neurological disorders, since human exposure to toxic metals is common from anthropogenic and natural sources, and toxic metals have damaging properties that are suspected to underlie many of these disorders. Questions remain, however, as to how toxic metals enter the nervous system, if one or combinations of metals are sufficient to precipitate disease, and how toxic metal exposure results in different patterns of neuronal and white matter loss. The hypothesis presented here is that damage to selective locus ceruleus neurons from toxic metals causes dysfunction of the blood-brain barrier. This allows circulating toxicants to enter astrocytes, from where they are transferred to, and damage, oligodendrocytes, and neurons. The type of neurological disorder that arises depends on (i) which locus ceruleus neurons are damaged, (ii) genetic variants that give rise to susceptibility to toxic metal uptake, cytotoxicity, or clearance, (iii) the age, frequency, and duration of toxicant exposure, and (iv) the uptake of various mixtures of toxic metals. Evidence supporting this hypothesis is presented, concentrating on studies that have examined the distribution of toxic metals in the human nervous system. Clinicopathological features shared between neurological disorders are listed that can be linked to toxic metals. Details are provided on how the hypothesis applies to multiple sclerosis and the major neurodegenerative disorders. Further avenues to explore the toxic metal hypothesis for neurological disorders are suggested. In conclusion, environmental toxic metals may play a part in several common neurological disorders. While further evidence to support this hypothesis is needed, to protect the nervous system it would be prudent to take steps to reduce environmental toxic metal pollution from industrial, mining, and manufacturing sources, and from the burning of fossil fuels.
Collapse
Affiliation(s)
- Roger Pamphlett
- Department of Pathology, Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - David P. Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
42
|
Ortega Moreno L, Bagues A, Martínez V, Abalo R. New Pieces for an Old Puzzle: Approaching Parkinson's Disease from Translatable Animal Models, Gut Microbiota Modulation, and Lipidomics. Nutrients 2023; 15:2775. [PMID: 37375679 DOI: 10.3390/nu15122775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Parkinson's disease (PD) is a severe neurodegenerative disease characterized by disabling motor alterations that are diagnosed at a relatively late stage in its development, and non-motor symptoms, including those affecting the gastrointestinal tract (mainly constipation), which start much earlier than the motor symptoms. Remarkably, current treatments only reduce motor symptoms, not without important drawbacks (relatively low efficiency and impactful side effects). Thus, new approaches are needed to halt PD progression and, possibly, to prevent its development, including new therapeutic strategies that target PD etiopathogeny and new biomarkers. Our aim was to review some of these new approaches. Although PD is complex and heterogeneous, compelling evidence suggests it might have a gastrointestinal origin, at least in a significant number of patients, and findings in recently developed animal models strongly support this hypothesis. Furthermore, the modulation of the gut microbiome, mainly through probiotics, is being tested to improve motor and non-motor symptoms and even to prevent PD. Finally, lipidomics has emerged as a useful tool to identify lipid biomarkers that may help analyze PD progression and treatment efficacy in a personalized manner, although, as of today, it has only scarcely been applied to monitor gut motility, dysbiosis, and probiotic effects in PD. Altogether, these new pieces should be helpful in solving the old puzzle of PD.
Collapse
Affiliation(s)
- Lorena Ortega Moreno
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
| | - Ana Bagues
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
| | - Vicente Martínez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Neuroscience Institute, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28049 Madrid, Spain
| | - Raquel Abalo
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
- Working Group of Basic Sciences on Pain and Analgesia of the Spanish Pain Society, 28046 Madrid, Spain
- Working Group of Basic Sciences on Cannabinoids of the Spanish Pain Society, 28046 Madrid, Spain
| |
Collapse
|
43
|
Ding Z, Fan X, Zhang Y, Yao M, Wang G, Dong Y, Liu J, Song W. The glymphatic system: a new perspective on brain diseases. Front Aging Neurosci 2023; 15:1179988. [PMID: 37396658 PMCID: PMC10308198 DOI: 10.3389/fnagi.2023.1179988] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/29/2023] [Indexed: 07/04/2023] Open
Abstract
The glymphatic system is a brain-wide perivascular pathway driven by aquaporin-4 on the endfeet of astrocytes, which can deliver nutrients and active substances to the brain parenchyma through periarterial cerebrospinal fluid (CSF) influx pathway and remove metabolic wastes through perivenous clearance routes. This paper summarizes the composition, overall fluid flow, solute transport, related diseases, affecting factors, and preclinical research methods of the glymphatic system. In doing so, we aim to provide direction and reference for more relevant researchers in the future.
Collapse
|
44
|
Whole CNS 3D Cryo-Fluorescence Tomography Shows CSF Clearance along Nasal Lymphatics, Spinal Nerves, and Lumbar/Sacral Lymph Nodes. J Imaging 2023; 9:jimaging9020045. [PMID: 36826964 PMCID: PMC9960470 DOI: 10.3390/jimaging9020045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Unwanted proteins and metabolic waste in cerebral spinal fluid are cleared from the brain by meningeal and nasal lymphatics and the perineural sheath of cranial nerves; however, the distribution and clearance of cerebral spinal fluid (CSF) along the subarachnoid space of the entire spinal cord is not fully understood. Cryo-fluorescence tomography (CFT) was used to follow the movement of tracers from the ventricular system of the brain down through the meningeal lining of the spinal cord and out to the spinal lymphatic nodes. Isoflurane-anesthetized mice were infused into the lateral cerebroventricle with 5.0 µL of quantum dots [QdotR 605 ITKTM amino (PEG)] over two mins. Mice were allowed to recover (ca 2-3 min) and remained awake and ambulatory for 5, 15, 30, 60, and 120 min after which they were euthanized, and the entire intact body was frozen at -80°. The entire mouse was sectioned, and white light and fluorescent images were captured after each slice to produce high resolution three-dimensional volumes. Tracer appeared throughout the ventricular system and central canal of the spinal cord and the entire subarachnoid space of the CNS. A signal could be visualized in the nasal cavity, deep cervical lymph nodes, thoracic lymph nodes, and more superficial submandibular lymph nodes as early as 15 min post infusion. A fluorescent signal could be visualized along the dorsal root ganglia and down the proximal extension of the spinal nerves of the thoracic and lumbar segments at 30 min. There was a significant accumulation of tracer in the lumbar and sacral lymph nodes between 15-60 min. The dense fluorescent signal in the thoracic vertebrae noted at 5- and 15-min post infusion was significantly reduced by 30 min. Indeed, all signals in the spinal cord were ostensibly absent by 120 min, except for trace amounts in the coccyx. The brain still had some residual signal at 120 min. These data show that Qdots with a hydrodynamic diameter of 16-20 nm rapidly clear from the brain of awake mice. These data also clearly demonstrate the rapid distribution and efflux of traces along a major length of the vertebral column and the potential contribution of the spinal cord in the clearance of brain waste.
Collapse
|