51
|
A multicentre validation study of the diagnostic value of plasma neurofilament light. Nat Commun 2021; 12:3400. [PMID: 34099648 PMCID: PMC8185001 DOI: 10.1038/s41467-021-23620-z] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Increased cerebrospinal fluid neurofilament light (NfL) is a recognized biomarker for neurodegeneration that can also be assessed in blood. Here, we investigate plasma NfL as a marker of neurodegeneration in 13 neurodegenerative disorders, Down syndrome, depression and cognitively unimpaired controls from two multicenter cohorts: King’s College London (n = 805) and the Swedish BioFINDER study (n = 1,464). Plasma NfL was significantly increased in all cortical neurodegenerative disorders, amyotrophic lateral sclerosis and atypical parkinsonian disorders. We demonstrate that plasma NfL is clinically useful in identifying atypical parkinsonian disorders in patients with parkinsonism, dementia in individuals with Down syndrome, dementia among psychiatric disorders, and frontotemporal dementia in patients with cognitive impairment. Data-driven cut-offs highlighted the fundamental importance of age-related clinical cut-offs for disorders with a younger age of onset. Finally, plasma NfL performs best when applied to indicate no underlying neurodegeneration, with low false positives, in all age-related cut-offs. Cerebrospinal fluid neurofilament light (NfL) is a biomarker for neurodegeneration that can also be assessed in blood. Here the authors show in a validation study the potential for plasma NfL as a biomarker for several neurodegenerative diseases.
Collapse
|
52
|
Fujita K, Peng S, Ma Y, Tang CC, Hellman M, Feigin A, Eidelberg D, Dhawan V. Blood-brain barrier permeability in Parkinson's disease patients with and without dyskinesia. J Neurol 2021; 268:2246-2255. [PMID: 33502551 PMCID: PMC11197155 DOI: 10.1007/s00415-021-10411-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/09/2020] [Accepted: 01/15/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Recent studies on a rodent model of Parkinson's disease (PD) have raised the possibility of increased blood-brain barrier (BBB) permeability, demonstrated by histology, autoradiography, and positron emission tomography (PET). However, in human PD patients, in vivo evidence of increased BBB permeability is lacking. We examined the hypothesis that levodopa treatment increases BBB permeability in human subjects with PD, particularly in those with levodopa-induced dyskinesia (LID). METHODS We used rubidium-82 (82Rb) and PET to quantify BBB influx in vivo in 19 PD patients, including eight with LID, and 12 age- and sex-matched healthy subjects. All subjects underwent baseline 82Rb scans. Seventeen chronically levodopa-treated patients were additionally rescanned during intravenous levodopa infusion. Influx rate constant, K1, by compartmental modeling or net influx transport, Ki, by graphical approach could not be estimated reliably. However, Vd, the "apparent volume of distribution" based on the 82Rb concentration in brain tissue and blood, was estimated with good stability as a local measure of the volume of distribution. RESULTS Rubidium influx into brain tissue was undetectable in PD patients with or without LID, scanned on and off drug. No significant differences in regional Vd were observed for PD patients with or without LID relative to healthy subjects, except in left thalamus. Moreover, changes in Vd measured off- and on-levodopa infusion were also not significant for dyskinetic and non-dyskinetic subjects. CONCLUSION 82Rb PET did not reveal significant changes in BBB permeability in PD patients.
Collapse
Affiliation(s)
- Koji Fujita
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shichun Peng
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Yilong Ma
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Matthew Hellman
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Andrew Feigin
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Vijay Dhawan
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
53
|
Eskildsen SF, Iranzo A, Stokholm MG, Stær K, Østergaard K, Serradell M, Otto M, Svendsen KB, Garrido A, Vilas D, Borghammer P, Santamaria J, Møller A, Gaig C, Brooks DJ, Tolosa E, Østergaard L, Pavese N. Impaired cerebral microcirculation in isolated REM sleep behaviour disorder. Brain 2021; 144:1498-1508. [PMID: 33880533 DOI: 10.1093/brain/awab054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/19/2020] [Accepted: 12/09/2020] [Indexed: 01/18/2023] Open
Abstract
During the prodromal period of Parkinson's disease and other α-synucleinopathy-related parkinsonisms, neurodegeneration is thought to progressively affect deep brain nuclei, such as the locus coeruleus, caudal raphe nucleus, substantia nigra, and the forebrain nucleus basalis of Meynert. Besides their involvement in the regulation of mood, sleep, behaviour, and memory functions, these nuclei also innervate parenchymal arterioles and capillaries throughout the cortex, possibly to ensure that oxygen supplies are adjusted according to the needs of neural activity. The aim of this study was to examine whether patients with isolated REM sleep behaviour disorder, a parasomnia considered to be a prodromal phenotype of α-synucleinopathies, reveal microvascular flow disturbances consistent with disrupted central blood flow control. We applied dynamic susceptibility contrast MRI to characterize the microscopic distribution of cerebral blood flow in the cortex of 20 polysomnographic-confirmed patients with isolated REM sleep behaviour disorder (17 males, age range: 54-77 years) and 25 healthy matched controls (25 males, age range: 58-76 years). Patients and controls were cognitively tested by Montreal Cognitive Assessment and Mini Mental State Examination. Results revealed profound hypoperfusion and microvascular flow disturbances throughout the cortex in patients compared to controls. In patients, the microvascular flow disturbances were seen in cortical areas associated with language comprehension, visual processing and recognition and were associated with impaired cognitive performance. We conclude that cortical blood flow abnormalities, possibly related to impaired neurogenic control, are present in patients with isolated REM sleep behaviour disorder and associated with cognitive dysfunction. We hypothesize that pharmacological restoration of perivascular neurotransmitter levels could help maintain cognitive function in patients with this prodromal phenotype of parkinsonism.
Collapse
Affiliation(s)
- Simon F Eskildsen
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alex Iranzo
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Morten G Stokholm
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Stær
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Karen Østergaard
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Mónica Serradell
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Marit Otto
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Alicia Garrido
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Dolores Vilas
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Per Borghammer
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Joan Santamaria
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Arne Møller
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Carles Gaig
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - David J Brooks
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Translational and Clinical Research Institute, Newcastle University, England, UK
| | - Eduardo Tolosa
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain.,Parkinson disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Neuroradiology Research Unit, Department of Radiology, Aarhus University Hospital, Denmark
| | - Nicola Pavese
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Translational and Clinical Research Institute, Newcastle University, England, UK
| |
Collapse
|
54
|
Pierzchlińska A, Kwaśniak-Butowska M, Sławek J, Droździk M, Białecka M. Arterial Blood Pressure Variability and Other Vascular Factors Contribution to the Cognitive Decline in Parkinson's Disease. Molecules 2021; 26:molecules26061523. [PMID: 33802165 PMCID: PMC8001922 DOI: 10.3390/molecules26061523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/24/2022] Open
Abstract
Dementia is one of the most disabling non-motor symptoms in Parkinson’s disease (PD). Unlike in Alzheimer’s disease, the vascular pathology in PD is less documented. Due to the uncertain role of commonly investigated metabolic or vascular factors, e.g., hypertension or diabetes, other factors corresponding to PD dementia have been proposed. Associated dysautonomia and dopaminergic treatment seem to have an impact on diurnal blood pressure (BP) variability, which may presumably contribute to white matter hyperintensities (WMH) development and cognitive decline. We aim to review possible vascular and metabolic factors: Renin-angiotensin-aldosterone system, vascular endothelial growth factor (VEGF), hyperhomocysteinemia (HHcy), as well as the dopaminergic treatment, in the etiopathogenesis of PD dementia. Additionally, we focus on the role of polymorphisms within the genes for catechol-O-methyltransferase (COMT), apolipoprotein E (APOE), vascular endothelial growth factor (VEGF), and for renin-angiotensin-aldosterone system components, and their contribution to cognitive decline in PD. Determining vascular risk factors and their contribution to the cognitive impairment in PD may result in screening, as well as preventive measures.
Collapse
Affiliation(s)
- Anna Pierzchlińska
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
- Correspondence: (A.P.); (M.D.)
| | - Magdalena Kwaśniak-Butowska
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Jarosław Sławek
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland
- Correspondence: (A.P.); (M.D.)
| | - Monika Białecka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
| |
Collapse
|
55
|
Weber CM, Clyne AM. Sex differences in the blood-brain barrier and neurodegenerative diseases. APL Bioeng 2021; 5:011509. [PMID: 33758788 PMCID: PMC7968933 DOI: 10.1063/5.0035610] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
The number of people diagnosed with neurodegenerative diseases is on the rise. Many of these diseases, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and motor neuron disease, demonstrate clear sexual dimorphisms. While sex as a biological variable must now be included in animal studies, sex is rarely included in in vitro models of human neurodegenerative disease. In this Review, we describe these sex-related differences in neurodegenerative diseases and the blood-brain barrier (BBB), whose dysfunction is linked to neurodegenerative disease development and progression. We explain potential mechanisms by which sex and sex hormones affect BBB integrity. Finally, we summarize current in vitro BBB bioengineered models and highlight their potential to study sex differences in BBB integrity and neurodegenerative disease.
Collapse
Affiliation(s)
- Callie M. Weber
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| |
Collapse
|
56
|
Wang T, Shi C, Luo H, Zheng H, Fan L, Tang M, Su Y, Yang J, Mao C, Xu Y. Neuroinflammation in Parkinson's Disease: Triggers, Mechanisms, and Immunotherapies. Neuroscientist 2021; 28:364-381. [PMID: 33576313 DOI: 10.1177/1073858421991066] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease involving multiple etiologies and pathogenesis, in which neuroinflammation is a common factor. Both preclinical experiments and clinical studies provide evidence for the involvement of neuroinflammation in the pathophysiology of PD, although there are a number of key issues related to neuroinflammatory processes in PD that remain to be addressed. In this review, we highlight the relationship between the common pathological mechanisms of PD and neuroinflammation, including aggregation of α-synuclein, genetic factors, mitochondrial dysfunction, and gut microbiome dysbiosis. We also describe the two positive feedback loops initiated in PD after the immune system is activated, and their role in the pathogenesis of PD. In addition, the interconnections and differences between the central and peripheral immune systems are discussed. Finally, we review the latest progress in immunotherapy research for PD patients, and propose future directions for clinical research.
Collapse
Affiliation(s)
- Tai Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
57
|
Elabi O, Gaceb A, Carlsson R, Padel T, Soylu-Kucharz R, Cortijo I, Li W, Li JY, Paul G. Human α-synuclein overexpression in a mouse model of Parkinson's disease leads to vascular pathology, blood brain barrier leakage and pericyte activation. Sci Rep 2021; 11:1120. [PMID: 33441868 PMCID: PMC7806665 DOI: 10.1038/s41598-020-80889-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
The pathological hallmark of Parkinson's disease (PD) is the formation of Lewy bodies containing aggregated alpha-synuclein (α-syn). Although PD is associated with these distinct histological changes, other pathological features such as microvascular alterations have been linked to neurodegeneration. These changes need to be investigated as they create a hostile brain microenvironment and may contribute to the development and progression of the disease. We use a human α-syn overexpression mouse model that recapitulates some of the pathological features of PD in terms of progressive aggregation of human α-syn, impaired striatal dopamine fiber density, and an age-dependent motor deficit consistent with an impaired dopamine release. We demonstrate for the first time in this model a compromised blood-brain barrier integrity and dynamic changes in vessel morphology from angiogenesis at earlier stages to vascular regression at later stages. The vascular alterations are accompanied by a pathological activation of pericytes already at an early stage without changing overall pericyte density. Our data support and further extend the occurrence of vascular pathology as an important pathophysiological aspect in PD. The model used provides a powerful tool to investigate disease-modifying factors in PD in a temporal sequence that might guide the development of new treatments.
Collapse
Affiliation(s)
- Osama Elabi
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden
| | - Thomas Padel
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden
| | - Rana Soylu-Kucharz
- Brain Disease Biomarker Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184, Lund, Sweden
| | - Irene Cortijo
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden
| | - Wen Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184, Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184, Lund, Sweden
- Institute of Health Sciences, China Medical University, Shenyang, 110122, China
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Sölvegatan 17, 22184, Lund, Sweden.
- Department of Neurology, Scania University Hospital, 22185, Lund, Sweden.
| |
Collapse
|
58
|
Al-Bachari S, Naish JH, Parker GJM, Emsley HCA, Parkes LM. Blood-Brain Barrier Leakage Is Increased in Parkinson's Disease. Front Physiol 2020; 11:593026. [PMID: 33414722 PMCID: PMC7784911 DOI: 10.3389/fphys.2020.593026] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
Background Blood–brain barrier (BBB) disruption has been noted in animal models of Parkinson’s disease (PD) and forms the basis of the vascular hypothesis of neurodegeneration, yet clinical studies are lacking. Objective To determine alterations in BBB integrity in PD, with comparison to cerebrovascular disease. Methods Dynamic contrast enhanced magnetic resonance images were collected from 49 PD patients, 15 control subjects with cerebrovascular disease [control positive (CP)] and 31 healthy control subjects [control negative (CN)], with all groups matched for age. Quantitative maps of the contrast agent transfer coefficient across the BBB (Ktrans) and plasma volume (vp) were produced using Patlak analysis. Differences in Ktrans and vp were assessed with voxel-based analysis as well as in regions associated with PD pathophysiology. In addition, the volume of white matter lesions (WMLs) was obtained from T2-weighted fluid attenuation inversion recovery (FLAIR) images. Results Higher Ktrans, reflecting higher BBB leakage, was found in the PD group than in the CN group using voxel-based analysis; differences were most prominent in the posterior white matter regions. Region of interest analysis confirmed Ktrans to be significantly higher in PD than in CN, predominantly driven by differences in the substantia nigra, normal-appearing white matter, WML and the posterior cortex. WML volume was significantly higher in PD compared to CN. Ktrans values and WML volume were similar in PD and CP, suggesting a similar burden of cerebrovascular disease despite lower cardiovascular risk factors. Conclusion These results show BBB disruption in PD.
Collapse
Affiliation(s)
- Sarah Al-Bachari
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom.,Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Josephine H Naish
- Division of Cardiovascular sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Bioxydyn Limited, Manchester, United Kingdom
| | - Geoff J M Parker
- Bioxydyn Limited, Manchester, United Kingdom.,Centre for Medical Image Computing, Department of Computer Science and Department of Neuroinflammation, University College London, London, United Kingdom
| | - Hedley C A Emsley
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom.,Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
59
|
Nissen SK, Ferreira SA, Nielsen MC, Schulte C, Shrivastava K, Hennig D, Etzerodt A, Graversen JH, Berg D, Maetzler W, Panhelainen A, Møller HJ, Brockmann K, Romero-Ramos M. Soluble CD163 Changes Indicate Monocyte Association With Cognitive Deficits in Parkinson's Disease. Mov Disord 2020; 36:963-976. [PMID: 33332647 PMCID: PMC8247308 DOI: 10.1002/mds.28424] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder with a significant immune component, as demonstrated by changes in immune biomarkers in patients' biofluids. However, which specific cells are responsible for those changes is unclear because most immune biomarkers can be produced by various cell types. OBJECTIVES The aim of this study was to explore monocyte involvement in PD. METHODS We investigated the monocyte-specific biomarker sCD163, the soluble form of the receptor CD163, in cerebrospinal fluid (CSF) and serum in two experiments, and compared it with other biomarkers and clinical data. Potential connections between CD163 and alpha-synuclein were studied in vitro. RESULTS CSF-sCD163 increased in late-stage PD and correlated with the PD biomarkers alpha-synuclein, Tau, and phosphorylated Tau, whereas it inversely correlated with the patients' cognitive scores, supporting monocyte involvement in neurodegeneration and cognition in PD. Serum-sCD163 increased only in female patients, suggesting a sex-distinctive monocyte response. CSF-sCD163 also correlated with molecules associated with adaptive and innate immune system activation and with immune cell recruitment to the brain. Serum-sCD163 correlated with proinflammatory cytokines and acute-phase proteins, suggesting a relation to chronic systemic inflammation. Our in vitro study showed that alpha-synuclein activates macrophages and induces shedding of sCD163, which in turn enhances alpha-synuclein uptake by myeloid cells, potentially participating in its clearance. CONCLUSIONS Our data present sCD163 as a potential cognition-related biomarker in PD and suggest a role for monocytes in both peripheral and brain immune responses. This may be directly related to alpha-synuclein's proinflammatory capacity but could also have consequences for alpha-synuclein processing. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | | | | | - Claudia Schulte
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research & German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | | | - Dorle Hennig
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Anders Etzerodt
- DANDRITE and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Daniela Berg
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research & German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts University, Kiel, Germany
| | - Walter Maetzler
- Department of Neurology, Christian-Albrechts University, Kiel, Germany
| | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Kathrin Brockmann
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research & German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | | |
Collapse
|
60
|
Paez AG, Gu C, Rajan S, Miao X, Cao D, Kamath V, Bakker A, Unschuld PG, Pantelyat AY, Rosenthal LS, Hua J. Differential Changes in Arteriolar Cerebral Blood Volume between Parkinson's Disease Patients with Normal and Impaired Cognition and Mild Cognitive Impairment (MCI) Patients without Movement Disorder - An Exploratory Study. Tomography 2020; 6:333-342. [PMID: 33364423 PMCID: PMC7744190 DOI: 10.18383/j.tom.2020.00033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cognitive impairment amongst Parkinson's disease (PD) patients is highly prevalent and associated with an increased risk of dementia. There is growing evidence that altered cerebrovascular functions contribute to cognitive impairment. Few studies have compared cerebrovascular changes in PD patients with normal and impaired cognition and those with mild-cognitive-impairment (MCI) without movement disorder. Here, we investigated arteriolar-cerebral-blood-volume (CBVa), an index reflecting the homeostasis of the most actively regulated segment in the microvasculature, using advanced MRI in various brain regions in PD and MCI patients and matched controls. Our goal is to find brain regions with altered CBVa that are specific to PD with normal and impaired cognition, and MCI-without-movement-disorder, respectively. In PD patients with normal cognition (n=10), CBVa was significantly decreased in the substantia nigra, caudate and putamen when compared to controls. In PD patients with impaired cognition (n=6), CBVa showed a decreasing trend in the substantia nigra, caudate and putamen, but was significantly increased in the presupplementary motor area and intracalcarine gyrus compared to controls. In MCI-patients-without-movement-disorder (n=18), CBVa was significantly increased in the caudate, putamen, hippocampus and lingual gyrus compared to controls. These findings provide important information for efforts towards developing biomarkers for the evaluation of potential risk of PD dementia (PDD) in PD patients. The current study is limited in sample size and therefore is exploratory in nature. The data from this pilot study will serve as the basis for power analysis for subsequent studies to further investigate and validate the current findings.
Collapse
Affiliation(s)
- Adrian G. Paez
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Chunming Gu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Suraj Rajan
- Department of Neurology; and
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Xinyuan Miao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| | - Di Cao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
- Department of Biomedical Engineering
| | - Vidyulata Kamath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; and
| | - Paul G. Unschuld
- Department of Psychogeriatric Medicine, Psychiatric University Hospital Zurich, Zurich, Switzerland
| | | | | | - Jun Hua
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
- Neurosection, Division of MR Research, Department of Radiology
| |
Collapse
|
61
|
Jeong JH, Ojha U, Lee YM. Pathological angiogenesis and inflammation in tissues. Arch Pharm Res 2020; 44:1-15. [PMID: 33230600 PMCID: PMC7682773 DOI: 10.1007/s12272-020-01287-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022]
Abstract
The role of angiogenesis in the growth of organs and tumors is widely recognized. Vascular-organ interaction is a key mechanism and a concept that enables an understanding of all biological phenomena and normal physiology that is essential for human survival under pathological conditions. Recently, vascular endothelial cells have been classified as a type of innate immune cells that are dependent on the pathological situations. Moreover, inflammatory cytokines and signaling regulators activated upon exposure to infection or various stresses play crucial roles in the pathological function of parenchymal cells, peripheral immune cells, stromal cells, and cancer cells in tissues. Therefore, vascular-organ interactions as a vascular microenvironment or tissue microenvironment under physiological and pathological conditions are gaining popularity as an interesting research topic. Here, we review vascular contribution as a major factor in microenvironment homeostasis in the pathogenesis of normal as well as cancerous tissues. Furthermore, we suggest that the normalization strategy of pathological angiogenesis could be a promising therapeutic target for various diseases, including cancer.
Collapse
Affiliation(s)
- Ji-Hak Jeong
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea.,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea
| | - You Mie Lee
- College of Pharmacy, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, Daegu, 41566, Republic of Korea. .,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
62
|
Yan Y, Yan Q, Qian L, Jiang Y, Chen X, Zeng S, Xu Z, Gong Z. S-adenosylmethionine administration inhibits levodopa-induced vascular endothelial growth factor-A expression. Aging (Albany NY) 2020; 12:21290-21307. [PMID: 33170152 PMCID: PMC7695432 DOI: 10.18632/aging.103863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Studies have demonstrated that S-adenosylmethionine could effectively affect the clinical wearing-off phenomena of levodopa, an antiparkinsonian agent; however, the detailed mechanisms for this effect need to be further clarified. RESULTS S-adenosylmethionine and levodopa had opposite effects on the protein stability of vascular endothelial growth factor-A. The analysis of tube formation and cell viability also showed the nonconforming functions of S-adenosylmethionine and levodopa on cell angiogenesis and proliferation. Meanwhile, S-adenosylmethionine could significantly abolish the increased angiogenesis and cell viability induced by levodopa. S-adenosylmethionine resulted in G1/S phase arrest, with decreased cyclin dependent kinase 4/6 and increased p16, a specific cyclin dependent kinase inhibitor. Mechanically, the different effects of levodopa and S-adenosylmethionine were dependent on the phosphorylation and activation of extracellular signal-regulated kinase. S-adenosylmethionine could be fitted into the predicted docking pocket in the crystal structure of vascular endothelial growth factor-A, enhancing its acetylation level and reducing half-life. CONCLUSIONS These observations suggested that methyl donor S-adenosylmethionine could act as a potential agent against vascular endothelial growth factor-A-related diseases induced by levodopa treatment. METHODS We performed in vitro cytological analyses to assess whether S-adenosylmethionine intake could influence levodopa-induced vascular endothelial growth factor-A expression in human umbilical vein endothelial cells.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Hunan, China
| | - Long Qian
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Hunan, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| |
Collapse
|
63
|
Ning B, Gu C, Guo G, Xu J, Bibic A, He X, Liu H, Chen L, Wei Z, Duan W, Liu P, Lu H, van Zijl PC, Ross CA, Smith W, Hua J. Mutant G2019S-LRRK2 Induces Abnormalities in Arteriolar Cerebral Blood Volume in Mouse Brains: An MRI Study. NEURODEGENER DIS 2020; 20:65-72. [PMID: 33152738 PMCID: PMC7864856 DOI: 10.1159/000510387] [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: 05/27/2020] [Accepted: 07/19/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease and the most common movement disorder characterized by motor impairments resulting from midbrain dopamine neuron loss. Abnormalities in small pial arteries and arterioles, which are the primary pathways of local delivery of nutrients and oxygen in brain tissue, have been reported in many neurodegenerative diseases including PD. Mutations in LRRK2 cause genetic PD and contribute to sporadic PD. The most common PD-linked mutation LRRK2 G2019S contributes 20-47% of genetic forms of PD in Caucasian populations. The human LRRK2 G2019S transgenic mouse model displays PD-like movement impairment and was used to identify novel LRRK2 inhibitors, which provides a useful model for studying microvascular abnormalities in PD. OBJECTIVES To investigate abnormalities in arteriolar cerebral blood volume (CBVa) in various brain regions using the inflow-based vascular-space occupancy (iVASO) MRI technique in LRRK2 mouse models of PD. METHODS Anatomical and iVASO MRI scans were performed in 5 female and 7 male nontransgenic (nTg), 3 female and 4 male wild-type (WT) LRRK2, and 5 female and 7 male G2019S-LRRK2 mice of 9 months of age. CBVa was calculated and compared in the substantia nigra (SN), olfactory cortex, and prefrontal cortex. RESULTS Compared to nTg mice, G2019S-LRRK2 mice showed decreased CBVa in the SN, but increased CBVa in the olfactory and prefrontal cortex in both male and female groups, whereas WT-LRRK2 mice showed no change in CBVa in the SN (male and female), the olfactory (female), and prefrontal (female) cortex, but a slight increase in CBVa in the olfactory and prefrontal cortex in the male group only. CONCLUSIONS Alterations in the blood volume of small arteries and arterioles (CBVa) were detected in the G2019S-LRRK2 mouse model of PD. The opposite changes in CBVa in the SN and the cortex indicate that PD pathology may have differential effects in different brain regions. Our results suggest the potential value of CBVa as a marker for clinical PD studies.
Collapse
Affiliation(s)
- Bo Ning
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
| | - Chunming Gu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Gongbo Guo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
| | - Jiadi Xu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Adnan Bibic
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Xiaofei He
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
| | - Hongshuai Liu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
| | - Lin Chen
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Zhiliang Wei
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Wenzhen Duan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peiying Liu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Peter C.M. van Zijl
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| | - Christopher A. Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wanli Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Univesity School of Medicine, Baltimore, Maryland, USA
| | - Jun Hua
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
| |
Collapse
|
64
|
Xiao M, Xiao ZJ, Yang B, Lan Z, Fang F. Blood-Brain Barrier: More Contributor to Disruption of Central Nervous System Homeostasis Than Victim in Neurological Disorders. Front Neurosci 2020; 14:764. [PMID: 32903669 PMCID: PMC7438939 DOI: 10.3389/fnins.2020.00764] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic but solid shield in the cerebral microvascular system. It plays a pivotal role in maintaining central nervous system (CNS) homeostasis by regulating the exchange of materials between the circulation and the brain and protects the neural tissue from neurotoxic components as well as pathogens. Here, we discuss the development of the BBB in physiological conditions and then focus on the role of the BBB in cerebrovascular disease, including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Finally, we summarize recent advancements in the development of therapies targeting the BBB and outline future directions and outstanding questions in the field. We propose that BBB dysfunction not only results from, but is causal in the pathogenesis of neurological disorders; the BBB is more a contributor to the disruption of CNS homeostasis than a victim in neurological disorders.
Collapse
Affiliation(s)
- Minjia Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Jie Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Yang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ziwei Lan
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Fang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
65
|
Vascular Inflammation Is a Risk Factor Associated with Brain Atrophy and Disease Severity in Parkinson's Disease: A Case-Control Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2591248. [PMID: 32733633 PMCID: PMC7376437 DOI: 10.1155/2020/2591248] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Abstract
Introduction Systemic inflammation with elevated oxidative stress causing neuroinflammation is considered a major factor in the pathogenesis of Parkinson's disease (PD). The interface between systemic circulation and the brain parenchyma is the blood-brain barrier (BBB), which also plays a role in maintaining neurovascular homeostasis. Vascular cell adhesion molecule-1 (VCAM-1) and microRNAs (miRNAs) regulate brain vessel endothelial function, neoangiogenesis, and, in turn, neuronal homeostasis regulation, such that their dysregulation can result in neurodegeneration, such as gray matter atrophy, in PD. Objective Our aim was to evaluate the associations among specific levels of gray matter atrophy, peripheral vascular adhesion molecules, miRNAs, and clinical disease severity in order to achieve a clearer understanding of PD pathogenesis. Methods Blood samples were collected from 33 patients with PD and 27 healthy volunteers, and the levels of VCAM-1 and several miRNAs in those samples were measured. Voxel-based morphometry (VBM) analysis was performed using 3 T magnetic resonance imaging (MRI) and SPM (Statistical Parametric Mapping software program). The associations among the vascular parameter, miRNAs, gray matter volume, and clinical disease severity measurements were evaluated by partial correlation analysis. Results The levels of VCAM-1, miRNA-22, and miRNA-29a expression were significantly elevated in the PD patients. The gray matter volume atrophy in the left parahippocampus, bilateral posterior cingulate gyrus, fusiform gyrus, left temporal gyrus, and cerebellum was significantly correlated with increased clinical disease severity, the upregulation of miRNA levels, and increased vascular inflammation. Conclusion Patients with PD seem to have abnormal levels of vascular inflammatory markers and miRNAs in the peripheral circulation, and these levels are correlated with specific brain volume changes. This study reinforces the associations among peripheral inflammation, the BBB interface, and gray matter atrophy in PD and further demonstrates that BBB dysfunction with neurovascular impairment may play an important role in PD progression.
Collapse
|
66
|
El-Ghaiesh SH, Bahr HI, Ibrahiem AT, Ghorab D, Alomar SY, Farag NE, Zaitone SA. Metformin Protects From Rotenone-Induced Nigrostriatal Neuronal Death in Adult Mice by Activating AMPK-FOXO3 Signaling and Mitigation of Angiogenesis. Front Mol Neurosci 2020; 13:84. [PMID: 32625061 PMCID: PMC7314970 DOI: 10.3389/fnmol.2020.00084] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease that affects substantia nigra dopamine neurons. Many studies have documented the role of oxidative stress and angiogenesis in the pathogenesis of PD. Metformin (MTF) is an antidiabetic medication and AMP-activated protein kinase (AMPK) regulator that has shown antioxidant and antiangiogenic properties in many disorders. The aim of this study is to investigate the neuroprotective effect of MTF in a mouse model of rotenone-prompted PD with a highlight on its influence on the AMPK/forkhead box transcription factor O3 (FOXO3) pathway and striatal angiogenesis. In the running study, PD was induced in mice using repeated doses of rotenone and concomitantly treated with MTF 100 or 200 mg/kg/day for 18 days. Rotarod and pole tests were used to examine the animals’ motor functionality. After that, animals were sacrificed, and brains were isolated and processed for immunohistochemical investigations or biochemical analyses. Oxidant stress and angiogenic markers were measured, including reduced glutathione, malondialdehyde, the nuclear factor erythroid 2–related factor 2 (Nrf2), hemoxygenase-1, thioredoxin, AMPK, FOXO3, and vascular endothelial growth factor (VEGF). Results indicated that MTF improved animals’ motor function, improved striatal glutathione, Nrf2, hemoxygenase-1, and thioredoxin. Furthermore, MTF upregulated AMPK-FOXO3 proteins and reduced VEGF and cleaved caspase 3. MTF also increased the number of tyrosine hydroxylase (TH)–stained neurons in the substantia nigra neurons and in striatal neuronal terminals. This study is the first to highlight that the neuroprotective role of MTF is mediated through activation of AMPK-FOXO3 signaling and inhibition of the proangiogenic factor, VEGF. Further studies are warranted to confirm this mechanism in other models of PD and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda I Bahr
- Department of Biochemistry, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Afaf T Ibrahiem
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Ghorab
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Suliman Y Alomar
- Doping Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Noha E Farag
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Department of Physiology, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
67
|
Tan EK, Chao YX, West A, Chan LL, Poewe W, Jankovic J. Parkinson disease and the immune system - associations, mechanisms and therapeutics. Nat Rev Neurol 2020; 16:303-318. [PMID: 32332985 DOI: 10.1038/s41582-020-0344-4] [Citation(s) in RCA: 292] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
Multiple lines of evidence indicate that immune system dysfunction has a role in Parkinson disease (PD); this evidence includes clinical and genetic associations between autoimmune disease and PD, impaired cellular and humoral immune responses in PD, imaging evidence of inflammatory cell activation and evidence of immune dysregulation in experimental models of PD. However, the mechanisms that link the immune system with PD remain unclear, and the temporal relationships of innate and adaptive immune responses with neurodegeneration are unknown. Despite these challenges, our current knowledge provides opportunities to develop immune-targeted therapeutic strategies for testing in PD, and clinical studies of some approaches are under way. In this Review, we provide an overview of the clinical observations, preclinical experiments and clinical studies that provide evidence for involvement of the immune system in PD and that help to define the nature of this association. We consider autoimmune mechanisms, central and peripheral inflammatory mechanisms and immunogenetic factors. We also discuss the use of this knowledge to develop immune-based therapeutic approaches, including immunotherapy that targets α-synuclein and the targeting of immune mediators such as inflammasomes. We also consider future research and clinical trials necessary to maximize the potential of targeting the immune system.
Collapse
Affiliation(s)
- Eng-King Tan
- Department of Neurology, Singapore General Hospital, Singapore, Singapore.
- National Neuroscience Institute, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| | - Yin-Xia Chao
- Department of Neurology, Singapore General Hospital, Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Andrew West
- Duke Center for Neurodegeneration and Neurotherapeutics, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Ling-Ling Chan
- Duke-NUS Medical School, Singapore, Singapore
- Department of Radiology, Singapore General Hospital, Singapore, Singapore
| | - Werner Poewe
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
68
|
Hirsch EC, Standaert DG. Ten Unsolved Questions About Neuroinflammation in Parkinson's Disease. Mov Disord 2020; 36:16-24. [PMID: 32357266 DOI: 10.1002/mds.28075] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease is a progressive and debilitating disorder that has so far eluded attempts to develop disease-modifying treatment. Both epidemiological and genetic studies support a role of neuroinflammation in the pathophysiology of Parkinson's disease. Postmortem studies and experimental analyses suggest the involvement of both innate and adaptive immunity in the degenerative process. There is also some circumstantial evidence for effects of immune therapies on the disease. In the present article, we review 10 unanswered questions related to neuroinflammatory processes in Parkinson's disease with the goal of stimulating research in the field and accelerating the clinical development of neuroprotective therapies based on anti-inflammatory strategies. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Etienne C Hirsch
- Faculté de Médecine de Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, U 1127, CNRS, Unité Mixte de Recherche (UMR) 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
69
|
Daneshvar Kakhaki R, Kouchaki E, Dadgostar E, Behnam M, Tamtaji OR, Nikoueinejad H, Akbari H. The correlation of helios and neuropilin-1 frequencies with parkinson disease severity. Clin Neurol Neurosurg 2020; 192:105833. [PMID: 32305590 DOI: 10.1016/j.clineuro.2020.105833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 02/11/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Parkinson disease (PD), a neurodegenerative disease, has also some immunologic basis in which several regulatory factors, like Helios and Neuropilin-1 (NRP-1) may show some roles in its pathogenesis. We aimed to evaluate the circulatory frequency of T regulatory cells (Tregs) expressing Helios and NRP-1 in PD. PATIENTS AND METHODS In this case-control study, 83 patients with PD and 83 healthy controls were enrolled. The diagnosis of PD was accomplished in accordance with clinical diagnostic criteria of the UK Parkinson Disease Society Brain Bank. The modified Hoehn and Yahr (H and Y) were used to measure the severity of PD. Flow cytometry was used to evaluate the circulatory frequency of CD4+CD25+Foxp3+Tregs expressing and Helios and NRP-1 in all participants. Also, correlation of H and Y with such frequencies was evaluated. RESULTS Our findings showed that frequency of CD4+CD25+Foxp3+Tregs expressing NRP-1 (P = 0.04) and Helios (P = 0.01) in patients with PD was significantly higher than those in healthy subjects. The frequency of Tregs expressing Helios and NRP-1 showed a negative correlation with H and Y criteria and disease duration. Multiple linear regression analysis indicated that the severity of PD is the only effective factor on the frequency of CD4+CD25+Foxp3+NRP-1+Tregs (P = 0.012) and CD4+CD25+FoxP3+ Helios + Tregs (P = 0.038). CONCLUSION Our study showed that the increased frequency of Tregs expressing Helios and NRP-1 is associated with the severity of PD.
Collapse
Affiliation(s)
- Reza Daneshvar Kakhaki
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Ebrahim Kouchaki
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Mohammad Behnam
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hassan Nikoueinejad
- Department of Immunology, Baqiyatallah University of Medical Sciences, Tehran, Iran; Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Baqiyatallah Hospital, Mollasadra Ave., Vanak Sq., P.O. Box: 19395-5487, Tehran, Iran.
| | - Hossein Akbari
- Department of Biostatistics and Public Health, Faculty of Health, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
70
|
The Link between Gut Dysbiosis and Neuroinflammation in Parkinson’s Disease. Neuroscience 2020; 432:160-173. [DOI: 10.1016/j.neuroscience.2020.02.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/16/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
|
71
|
Meng XY, Huang AQ, Khan A, Zhang L, Sun XQ, Song H, Han J, Sun QR, Wang YD, Li XL. Vascular endothelial growth factor-loaded poly-lactic-co-glycolic acid nanoparticles with controlled release protect the dopaminergic neurons in Parkinson's rats. Chem Biol Drug Des 2020; 95:631-639. [PMID: 32167672 DOI: 10.1111/cbdd.13681] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/11/2019] [Accepted: 11/24/2019] [Indexed: 12/31/2022]
Abstract
Vascular endothelial growth factor (VEGF) had neuroprotective effects on dopaminergic (DA) neurons. In order to overcome the gastrointestinal digestion and bioaccessibility, VEGF was encapsulated with poly-lactic-co-glycolic acid nanospheres (NS) in order to prevent the VEGF degradation until its release. The caudal administration of VEGF and NS encapsulated VEGF at different doses (1.0, 10.0, and 100.0 ng/ml) on the rats with Parkinson's disease lesion was evaluated. Intravenous injected VEGF at the dose of 1 ng/ml displayed the strongest neuroprotective effect than other groups as well as the stereotaxic group. The NS encapsulated with VEGF can pass through blood-brain barrier and protect the DA neurons. There was no significant difference between intravenous injection method and stereotaxic method, while the first method is simpler and convenient. Injection of NS encapsulated with VEGF may become a valuable neurorescuing therapeutic approach for Parkinson's disease.
Collapse
Affiliation(s)
- Xian-Yue Meng
- Department of Neurology, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| | - An-Qi Huang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Li Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| | - Xiao-Qian Sun
- Department of Neurology, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| | - Hao Song
- Centre for Stem Cell and Regenerative Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Jun Han
- College of Pharmacy, Liaocheng University, Liaocheng, China
| | - Qian-Ru Sun
- Department of Neuroimmune Laboratory, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| | - Yu-Dan Wang
- Engineering Research Center of Biopolymer Functional Materials of Yunnan, Yunnan Minzu University, Kunming, China
| | - Xue-Li Li
- Department of Neurology, Liaocheng People's Hospital and Liaocheng School of Clinical Medicine, Taishan Medical University, Liaocheng, China
| |
Collapse
|
72
|
Donaghy PC, Firbank M, Mitra D, Petrides G, Lloyd J, Barnett N, Olsen K, Thomas AJ, O'Brien JT. Microbleeds in dementia with Lewy bodies. J Neurol 2020; 267:1491-1498. [PMID: 32016624 PMCID: PMC7184053 DOI: 10.1007/s00415-020-09736-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Microbleeds are associated with the development of dementia in older people and are common in Alzheimer's disease (AD). Their prevalence and clinical importance in dementia with Lewy bodies (DLB) is unclear. The objective of this study was to compare the rates of microbleeds in DLB with those in AD and healthy older people, and investigate associations between microbleeds and amyloid deposition, vascular risk and disease severity in DLB. METHODS DLB (n = 30), AD (n = 18) and control (n = 20) participants underwent clinical assessment at baseline and 1 year in this longitudinal observational study. 3T MRI (including T2* susceptibility weighted imaging) and florbetapir PET were carried out at baseline. Microbleeds were rated visually and a standardised uptake value ratio (SUVR) was calculated from florbetapir PET scans. RESULTS 40% of DLB subjects had microbleeds compared with 50% of AD and 15% of controls. Compared to DLB without microbleeds, those with microbleeds had higher systolic BP (156 ± 26 v. 135 ± 19 mmHg; p = 0.03), but did not have greater levels of vascular disease or amyloid deposition (SUVR 1.25 ± 0.24 v. 1.25 ± 0.22; p = 0.33). There was evidence of less severe dementia in DLB participants with microbleeds, but these differences may have been driven by a shorter disease duration in those with microbleeds. CONCLUSION The presence of microbleeds in DLB is associated with higher blood pressure, but not with other measures of vascular disease or amyloid deposition. The relationship between microbleeds and clinical presentation remains unclear.
Collapse
Affiliation(s)
- Paul C Donaghy
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Michael Firbank
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Dipayan Mitra
- Neuroradiology Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - George Petrides
- Nuclear Medicine Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jim Lloyd
- Nuclear Medicine Department, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nicola Barnett
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kirsty Olsen
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Alan J Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
73
|
Kim S, Lee M, Choi YK. The Role of a Neurovascular Signaling Pathway Involving Hypoxia-Inducible Factor and Notch in the Function of the Central Nervous System. Biomol Ther (Seoul) 2020; 28:45-57. [PMID: 31484285 PMCID: PMC6939687 DOI: 10.4062/biomolther.2019.119] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
In the neurovascular unit, the neuronal and vascular systems communicate with each other. O2 and nutrients, reaching endothelial cells (ECs) through the blood stream, spread into neighboring cells, such as neural stem cells, and neurons. The proper function of neural circuits in adults requires sufficient O2 and glucose for their metabolic demands through angiogenesis. In a central nervous system (CNS) injury, such as glioma, Parkinson’s disease, and Alzheimer’s disease, damaged ECs can contribute to tissue hypoxia and to the consequent disruption of neuronal functions and accelerated neurodegeneration. This review discusses the current evidence regarding the contribution of oxygen deprivation to CNS injury, with an emphasis on hypoxia-inducible factor (HIF)-mediated pathways and Notch signaling. Additionally, it focuses on adult neurological functions and angiogenesis, as well as pathological conditions in the CNS. Furthermore, the functional interplay between HIFs and Notch is demonstrated in pathophysiological conditions.
Collapse
Affiliation(s)
- Seunghee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Minjae Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
74
|
Boi L, Pisanu A, Greig NH, Scerba MT, Tweedie D, Mulas G, Fenu S, Carboni E, Spiga S, Carta AR. Immunomodulatory drugs alleviate l-dopa-induced dyskinesia in a rat model of Parkinson's disease. Mov Disord 2019; 34:1818-1830. [PMID: 31335998 PMCID: PMC11719776 DOI: 10.1002/mds.27799] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/05/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Thalidomide and closely related analogues are used clinically for their immunomodulatory and antiangiogenic properties mediated by the inhibition of the proinflammatory cytokine tumor necrosis factor α. Neuroinflammation and angiogenesis contribute to classical neuronal mechanisms underpinning the pathophysiology of l-dopa-induced dyskinesia, a motor complication associated with l-dopa therapy in Parkinson's disease. The efficacy of thalidomide and the more potent derivative 3,6'-dithiothalidomide on dyskinesia was tested in the 6-hydroxydopamine Parkinson's disease model. METHODS Three weeks after 6-hydroxydopamine infusion, rats received 10 days of treatment with l-dopa plus benserazide (6 mg/kg each) and thalidomide (70 mg/kg) or 3,6'-dithiothalidomide (56 mg/kg), and dyskinesia and contralateral turning were recorded daily. Rats were euthanized 1 hour after the last l-dopa injection, and levels of tumor necrosis factor-α, interleukin-10, OX-42, vimentin, and vascular endothelial growth factor immunoreactivity were measured in their striatum and substantia nigra reticulata to evaluate neuroinflammation and angiogenesis. Striatal levels of GLUR1 were measured as a l-dopa-induced postsynaptic change that is under tumor necrosis factor-α control. RESULTS Thalidomide and 3,6'-dithiothalidomide significantly attenuated the severity of l-dopa-induced dyskinesia while not affecting contralateral turning. Moreover, both compounds inhibited the l-dopa-induced microgliosis and excessive tumor necrosis factor-α in the striatum and substantia nigra reticulata, while restoring physiological levels of the anti-inflammatory cytokine interleukin-10. l-Dopa-induced angiogenesis was inhibited in both basal ganglia nuclei, and l-dopa-induced GLUR1 overexpression in the dorsolateral striatum was restored to normal levels. CONCLUSIONS These data suggest that decreasing tumor necrosis factor-α levels may be useful to reduce the appearance of dyskinesia, and thalidomide, and more potent derivatives may provide an effective therapeutic approach to dyskinesia. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Augusta Pisanu
- CNR Institute of Neuroscience, Cagliari, Cagliari, Italy
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Michael T. Scerba
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Sandro Fenu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
75
|
Kummer BR, Diaz I, Wu X, Aaroe AE, Chen ML, Iadecola C, Kamel H, Navi BB. Associations between cerebrovascular risk factors and parkinson disease. Ann Neurol 2019; 86:572-581. [PMID: 31464350 DOI: 10.1002/ana.25564] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To determine whether cerebrovascular risk factors are associated with subsequent diagnoses of Parkinson disease, and whether these associations are similar in magnitude to those with subsequent diagnoses of Alzheimer disease. METHODS This was a retrospective cohort study using claims data from a 5% random sample of Medicare beneficiaries from 2008 to 2015. The exposures were stroke, atrial fibrillation, coronary disease, hyperlipidemia, hypertension, sleep apnea, diabetes mellitus, heart failure, peripheral vascular disease, chronic kidney disease, chronic obstructive pulmonary disease, valvular heart disease, tobacco use, and alcohol abuse. The primary outcome was a new diagnosis of idiopathic Parkinson disease. The secondary outcome was a new diagnosis of Alzheimer disease. Marginal structural Cox models adjusting for time-dependent confounding were used to characterize the association between exposures and outcomes. We also evaluated the association between cerebrovascular risk factors and subsequent renal colic (negative control). RESULTS Among 1,035,536 Medicare beneficiaries followed for a mean of 5.2 years, 15,531 (1.5%) participants were diagnosed with Parkinson disease and 81,974 (7.9%) were diagnosed with Alzheimer disease. Most evaluated cerebrovascular risk factors, including prior stroke (hazard ratio = 1.55; 95% confidence interval = 1.39-1.72), were associated with the subsequent diagnosis of Parkinson disease. The magnitudes of these associations were similar, but attenuated, to the associations between cerebrovascular risk factors and Alzheimer disease. Confirming the validity of our analytical model, most cerebrovascular risk factors were not associated with the subsequent diagnosis of renal colic. INTERPRETATION Cerebrovascular risk factors are associated with Parkinson disease, an effect comparable to their association with Alzheimer disease. ANN NEUROL 2019;86:572-581.
Collapse
Affiliation(s)
- Benjamin R Kummer
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Iván Diaz
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY
| | - Xian Wu
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY
| | - Ashley E Aaroe
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Monica L Chen
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Babak B Navi
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
76
|
Bogorad MI, DeStefano JG, Linville RM, Wong AD, Searson PC. Cerebrovascular plasticity: Processes that lead to changes in the architecture of brain microvessels. J Cereb Blood Flow Metab 2019; 39:1413-1432. [PMID: 31208241 PMCID: PMC6681538 DOI: 10.1177/0271678x19855875] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The metabolic demands of the brain are met by oxygen and glucose, supplied by a complex hierarchical network of microvessels (arterioles, capillaries, and venules). Transient changes in neural activity are accommodated by local dilation of arterioles or capillaries to increase cerebral blood flow and hence nutrient availability. Transport and communication between the circulation and the brain is regulated by the brain microvascular endothelial cells that form the blood-brain barrier. Under homeostatic conditions, there is very little turnover in brain microvascular endothelial cells, and the cerebrovascular architecture is largely static. However, changes in the brain microenvironment, due to environmental factors, disease, or trauma, can result in additive or subtractive changes in cerebrovascular architecture. Additions occur by angiogenesis or vasculogenesis, whereas subtractions occur by vascular pruning, injury, or endothelial cell death. Here we review the various processes that lead to changes in the cerebrovascular architecture, including sustained changes in the brain microenvironment, development and aging, and injury, disease, and repair.
Collapse
Affiliation(s)
- Max I Bogorad
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jackson G DeStefano
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,3 Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Wong
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- 1 Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.,3 Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
77
|
Lindholm B, Eek F, Skogar Ö, Hansson EE. Dyskinesia and FAB score predict future falling in Parkinson's disease. Acta Neurol Scand 2019; 139:512-518. [PMID: 30820932 DOI: 10.1111/ane.13084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/09/2019] [Accepted: 02/26/2019] [Indexed: 12/11/2022]
Abstract
A growing body of research highlights the importance of cognition for prediction of falls in Parkinson's disease (PD). However, a previously proposed prediction model for future near falls and falls in PD, which includes history of near falls, tandem gait, and retropulsion, was developed without considering cognitive impairment. Therefore, by using a sample of 64 individuals with relatively mild PD and not excluding those with impaired cognition we aimed to externally validate the previously proposed model as well as to explore the value of additional predictors that also consider cognitive impairment. Since this validation study failed to support the proposed model in a PD sample including individuals with impaired global cognition, extended analyses generated a new model including dyskinesia (item 32 of Unified PD Rating Scale) and frontal lobe impairment (Frontal Assessment Battery-FAB) as significant independent predictors for future near falls and falls in PD. The discriminant ability of this new model was acceptable (AUC, 0. 80; 95% CI 0.68-0.91). Replacing the continuous FAB scores by a dichotomized version of FAB with a cut-off score ≤14 yielded slightly lower but still acceptable discriminant ability (AUC, 0. 79; 95% CI 0.68-0.91). Further studies are needed to test our new model and the proposed cut-off score of FAB in additional samples. Taken together, our observations suggest potentially important additions to the evidence base for clinical fall prediction in PD with concomitant cognitive impairment.
Collapse
Affiliation(s)
- Beata Lindholm
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö Lund University Lund Sweden
- Department of Neurology and Rehabilitation Medicine Skåne University Hospital Malmö Sweden
| | - Frida Eek
- Department of Health Sciences Lund University Lund Sweden
| | - Örjan Skogar
- Department of Neurobiology, Care Sciences and Society Karolinska Institutet Stockholm Sweden
- Institute of Gerontology and Aging Research Network – Jönköping (ARN‐J), School of Health and Welfare Jönköping University Jönköping Sweden
| | - Eva E. Hansson
- Department of Health Sciences Lund University Lund Sweden
| |
Collapse
|
78
|
Parkinson's and Lewy body dementia CSF biomarkers. Clin Chim Acta 2019; 495:318-325. [PMID: 31051162 DOI: 10.1016/j.cca.2019.04.078] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 11/24/2022]
Abstract
The clinical diagnosis of Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) is challenging due to highly variable clinical presentation and clinical and pathological overlap with other neurodegenerative diseases. Since cerebrospinal fluid (CSF) mirrors the pathological changes taking place in the brain, it represents a promising source of biomarkers. With respect to classical AD biomarkers, low CSF Aβ42 levels have shown a robust prognostic value in terms of development of cognitive impairment in PD and DLB. In the differential diagnosis between AD and DLB, a potential role of t-tau, p-tau and Aβ42/Aβ38 ratio has been demonstrated. Regarding CSF α-synuclein (α-syn) species, lower levels of total α-synuclein (t-α-syn) and higher concentration of oligomeric-α-synuclein (o-α-syn) and phosphorylated α-synuclein (p-α-syn) have been observed in PD. Furthermore, the detection of "pro-aggregating" α-synuclein has enabled the discrimination of patients affected by synucleinopathies with high sensitivity and specificity. New promising biomarkers are emerging: GCase activity (reduced in PD and DLB patients vs. controls), CSF/serum albumin ratio (increased in PD and DLB), fatty-acid-binding protein (increased in AD and DLB vs. PD), visinin-like protein-1 (increased in AD vs. DLB) and monoamines (useful in differential diagnosis among PD and DLB). These encouraging results need to be confirmed by future studies.
Collapse
|
79
|
Hansson O, Santillo AF, Meeter LH, Nilsson K, Landqvist Waldö M, Nilsson C, Blennow K, van Swieten JC, Janelidze S. CSF placental growth factor - a novel candidate biomarker of frontotemporal dementia. Ann Clin Transl Neurol 2019; 6:863-872. [PMID: 31139684 PMCID: PMC6529985 DOI: 10.1002/acn3.763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Objective Diagnosis of frontotemporal dementia (FTD) is complicated by the overlap of clinical symptoms with other dementia disorders. Development of robust fluid biomarkers is critical to improve the diagnostic work‐up of FTD. Methods CSF concentrations of placental growth factor (PlGF) were measured in the discovery cohort including patients with FTD (n = 27), Alzheimer disease (AD) dementia (n = 75), DLB or PDD (n = 47), subcortical vascular dementia (VaD, n = 33), mild cognitive impairment that later converted to AD (MCI‐AD, n = 34), stable MCI (sMCI, n = 62), and 50 cognitively healthy controls from the Swedish BioFINDER study. For validation, CSF PlGF was measured in additional independent cohort of FTD patients (n = 22) and controls (n = 18) from the Netherlands. Results In the discovery cohort, MCI, MCI‐AD, AD dementia, DLB‐PDD, VaD, and FTD patients all showed increased CSF levels of PlGF compared with controls (sMCI P = 0.019; MCI‐AD P = 0.005; AD dementia, DLB‐PDD, VaD, and FTD all P < 0.001). PlGF levels were 1.8–2.1‐fold higher in FTD than in AD, DLB‐PDD and VaD (all P < 0.001). PlGF distinguished with high accuracy FTD from controls and sMCI performing better than tau/Aβ42 (AUC 0.954–0.996 versus 0.564–0.754, P < 0.001). A combination of PlGF, tau, and Aβ42 (tau/Aβ42/PlGF) was more accurate than tau/Aβ42 when differentiating FTD from a group of other dementias (AUC 0.972 vs. 0.932, P < 0.01). Increased CSF levels of PlGF in FTD compared with controls were corroborated in the validation cohort. Interpretation CSF PlGF is increased in FTD compared with other dementia disorders, MCI, and healthy controls and might be useful as a diagnostic biomarker of FTD.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Memory Clinic Skåne University Hospital Malmö Sweden
| | - Alexander F Santillo
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Memory Clinic Skåne University Hospital Malmö Sweden
| | - Lieke H Meeter
- Department of Neurology Erasmus Medical Center Rotterdam The Netherlands
| | - Karin Nilsson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden
| | - Maria Landqvist Waldö
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Clinical Sciences Helsingborg Department of Clinical Sciences Lund University Lund Sweden
| | - Christer Nilsson
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden.,Department of Neurology Skåne University Hospital Lund Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry The Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden.,Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
| | - John C van Swieten
- Department of Neurology Erasmus Medical Center Rotterdam The Netherlands.,Department of Clinical Genetics VU University Medical Center Amsterdam The Netherlands
| | - Shorena Janelidze
- Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Malmö Sweden
| |
Collapse
|
80
|
Fuzzati-Armentero MT, Cerri S, Blandini F. Peripheral-Central Neuroimmune Crosstalk in Parkinson's Disease: What Do Patients and Animal Models Tell Us? Front Neurol 2019; 10:232. [PMID: 30941089 PMCID: PMC6433876 DOI: 10.3389/fneur.2019.00232] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is no longer considered an immune privileged organ and neuroinflammation has long been associated with Parkinson's disease. Accumulating evidence demonstrates that innate and adaptive responses take place in the CNS. The extent to which peripheral immune alterations impacts on the CNS, or vice and versa, is, however, still a matter of debate. Gaining a better knowledge of the molecular and cellular immune dysfunctions present in these two compartments and clarifying their mutual interactions is a fundamental step in understanding and preventing Parkinson's disease (PD) pathogenesis. This review provides an overview of the current knowledge on inflammatory processes evidenced both in PD patients and in toxin-induced animal models of the disease. It discusses differences and similarities between human and animal studies in the context of neuroinflammation and immune responses and how they have guided therapeutic strategies to slow down disease progression. Future longitudinal studies are necessary and can help gain a better understanding on peripheral-central nervous system crosstalk to improve therapeutic strategies for PD.
Collapse
|
81
|
Colín-Castelán D, Zaina S. Associations between atherosclerosis and neurological diseases, beyond ischemia-induced cerebral damage. Rev Endocr Metab Disord 2019; 20:15-25. [PMID: 30891682 DOI: 10.1007/s11154-019-09486-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neurodegeneration is traditionally viewed as a consequence of peptide accumulation in the brain, stroke and/or cerebral ischemia. Nonetheless, a number of scattered observations suggest that neurological disease and atherosclerosis may be linked by more complex mechanisms. Understanding the intricate link between atherosclerosis and neurological conditions may have a significant impact on the quality of life of the growing ageing population and of high cardiovascular risk groups in general. Epidemiological data support the notion that neurological dysfunction and atherosclerosis coexist long before any evident clinical complications of cardiovascular disease appear and may be causally linked. Baffling, often overlooked, molecular data suggest that nervous tissue-specific gene expression is relaxed specifically in the atheromatous vascular wall, and/or that a systemic dysregulation of genes involved in nervous system biology dictates a concomitant progression of neurological disease and atherosclerosis. Further epidemiological and experimental work is needed to clarify the details and clinical relevance of those complex links.
Collapse
Affiliation(s)
- Dannia Colín-Castelán
- Department of Medical Sciences, Division of Health Sciences, Campus León, University of Guanajuato, León, Guanajuato, Mexico.
| | - Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Campus León, University of Guanajuato, León, Guanajuato, Mexico
| |
Collapse
|
82
|
Garretti F, Agalliu D, Lindestam Arlehamn CS, Sette A, Sulzer D. Autoimmunity in Parkinson's Disease: The Role of α-Synuclein-Specific T Cells. Front Immunol 2019; 10:303. [PMID: 30858851 PMCID: PMC6397885 DOI: 10.3389/fimmu.2019.00303] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 12/23/2022] Open
Abstract
Evidence from a variety of studies implicates a role for the adaptive immune system in Parkinson's disease (PD). Similar to multiple sclerosis (MS) patients who display a high number of T cells in the brain attacking oligodendrocytes, PD patients show higher numbers of T cells in the ventral midbrain than healthy, age-matched controls. Mouse models of the disease also show the presence of T cells in the brain. The role of these infiltrating T cells in the propagation of disease is controversial; however, recent studies indicate that they may be autoreactive in nature, recognizing disease-altered self-proteins as foreign antigens. T cells of PD patients can generate an autoimmune response to α-synuclein, a protein that is aggregated in PD. α-Synuclein and other proteins are post-translationally modified in an environment in which protein processing is altered, possibly leading to the generation of neo-epitopes, or self-peptides that have not been identified by the host immune system as non-foreign. Infiltrating T cells may also be responding to such modified proteins. Genome-wide association studies (GWAS) have shown associations of PD with haplotypes of major histocompatibility complex (MHC) class II genes, and a polymorphism in a non-coding region that may increase MHC class II in PD patients. We speculate that the inflammation observed in PD may play both pathogenic and protective roles. Future studies on the adaptive immune system in neurodegenerative disorders may elucidate steps in disease pathogenesis and assist with the development of both biomarkers and treatments.
Collapse
Affiliation(s)
- Francesca Garretti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Pharmacology, Columbia University Irving Medical Center, New York, NY, United States.,Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
83
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1410] [Impact Index Per Article: 235.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
84
|
Kouchaki E, Daneshvar Kakhaki R, Tamtaji OR, Dadgostar E, Behnam M, Zaribaf A, Nikoueinejad H, Akbari H, Asemi Z. Correlation of serum levels and gene expression of tumor necrosis factor-α-induced protein-8 like-2 with Parkinson disease severity. Metab Brain Dis 2018; 33:1955-1959. [PMID: 30105613 DOI: 10.1007/s11011-018-0302-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023]
Abstract
Different immune-mediated mechanisms involved in the pathogenesis of Parkinson disease (PD) as a neurodegenerative and inflammatory disease. According to our knowledge, there is no report evaluating Tumor necrosis factor-α-induced protein-8 like-2 (TIPE2), a cytokine maintaining immune homeostasis, in PD. We analyzed the correlation of the serum levels and circulatory gene expression of TIPE2 with severity of PD. In this case-control study, 43 patients with PD and 40 healthy subjects were enrolled. The diagnosis of PD was performed byclinical diagnostic criteria of the UK Parkinson's Disease Society Brain Bank. The severity of PD was evaluated by modified Hoehn and Yahr (H and Y) scale. Serum levels and gene expression of TIPE2 were assessed by Elisa and real time PCR, respectively. The mean serum levels and gene expression of TIPE2 in patients with PD did not have significant difference compared to healthy subjects. Linear multiple regression analysis showed that increased serum levels of TIPE2 are positively related to age and severity of PD (P ≤ 0.0001). In addition, the gene expression of TIPE2 was found to be associated with age (P < 0.0001). Our study showed that the serum levels of TIPE2 and its gene expression might be important prognostic biomarkers of PD.
Collapse
Affiliation(s)
- Ebrahim Kouchaki
- Department of Neurology, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Omid Reza Tamtaji
- Halal Research Center of IRI, FDA, Tehran, Iran
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, I.R., Iran
| | - Ehsan Dadgostar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mohammad Behnam
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Zaribaf
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Hassan Nikoueinejad
- Department of Immunology, Baqiyatallah University of Medical Sciences, Tehran, Iran.
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Baqiyatallah Hospital, Mollasadra Ave., Vanak Sq, P.O. Box: 19395-5487, Tehran, I.R, Iran.
| | - Hossein Akbari
- Department of Biostatistics and Public Health, Faculty of Health, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R., Iran
| |
Collapse
|
85
|
Chen X, Hu Y, Cao Z, Liu Q, Cheng Y. Cerebrospinal Fluid Inflammatory Cytokine Aberrations in Alzheimer's Disease, Parkinson's Disease and Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Front Immunol 2018; 9:2122. [PMID: 30283455 PMCID: PMC6156158 DOI: 10.3389/fimmu.2018.02122] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/28/2018] [Indexed: 12/14/2022] Open
Abstract
It has been suggested that cytokine-mediated inflammation plays a key role for the onset and/or development of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). However, clinical studies have yielded inconsistent results for the aberrant cytokine levels in circulation of patients with AD, PD, and ALS. Previous studies have used meta-analysis to address the inconsistent data for blood cytokine levels in the patients with AD, PD, and ALS. Here, we performed a systemic review of cerebrospinal fluid inflammatory cytokine data in patients with AD, PD and ALS, and sought to quantitatively summarize the CSF inflammatory cytokine data with a meta-analytical technique. The systematic search from Pubmed and Web of Science identified 71 articles with 2629 patients and 2049 controls for the meta-analysis. Random-effects meta-analysis demonstrated that CSF TGF-β, MCP-1, and YKL-40 levels were significantly elevated in AD patients when compared with controls. In addition, patients with PD had heightened levels of TGF-β1, IL-6, and IL-1β in CSF. Furthermore, G-CSF, IL-2, IL-15, IL-17, MCP-1, MIP-1α, TNF-α, and VEGF levels were significantly increased in patients with ALS as compared with controls. Taken together, these results not only strengthen the clinical evidence that neurodegenerative diseases are accompanied by the increased inflammatory response, but also reveal the unique inflammatory response profile in the central nervous system of patients with AD, PD and ALS. Given the robust associations between some cytokines and neurodegenerative diseases found in this meta-analysis, CSF inflammatory cytokines may be used as biomarkers for these diseases in the future.
Collapse
Affiliation(s)
| | | | | | - Qingshan Liu
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| |
Collapse
|
86
|
Cerebrospinal fluid concentrations of inflammatory markers in Parkinson's disease and atypical parkinsonian disorders. Sci Rep 2018; 8:13276. [PMID: 30185816 PMCID: PMC6125576 DOI: 10.1038/s41598-018-31517-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/20/2018] [Indexed: 11/13/2022] Open
Abstract
Inflammation has been implicated in the pathogenesis of Parkinson’s disease (PD). We here investigate levels of inflammatory biomarkers in cerebrospinal fluid (CSF) in PD and atypical parkinsonian disorders (APD) compared with neurologically healthy controls. We included 131 patients with PD and 27 PD with dementia (PDD), 24 with multiple system atrophy (MSA), 14 with progressive supranuclear palsy (PSP) and 50 controls, all part of the Swedish BioFINDER study. CSF was analyzed for CRP, SAA, IL-6, IL-8, YKL-40 and MCP-1 (CCL2) as well as α-synuclein (α-syn), tau, tau phosphorylated at Thr181 (P-tau), Aβ42 and NfL. In this exploratory study, we found higher levels of the inflammatory biomarker SAA in PDD and MSA compared with controls and PD and higher levels of CRP in PDD and MSA compared with PD. YKL-40 was lower in PD compared with controls. There were multiple positive correlations between the inflammatory markers, α-syn and markers of neuroaxonal injury (NfL and tau). In PD, higher levels of inflammatory biomarkers correlated with worse motor function and cognitive impairment. Thus, inflammatory biomarkers were increased in PDD and MSA. Furthermore, inflammatory biomarkers correlated with more severe disease regarding motor symptoms and cognitive impairment in PD, indicating an association between inflammation and more aggressive disease course. However, the results need confirmation in follow-up studies.
Collapse
|
87
|
Bowman GL, Dayon L, Kirkland R, Wojcik J, Peyratout G, Severin IC, Henry H, Oikonomidi A, Migliavacca E, Bacher M, Popp J. Blood‐brain barrier breakdown, neuroinflammation, and cognitive decline in older adults. Alzheimers Dement 2018; 14:1640-1650. [DOI: 10.1016/j.jalz.2018.06.2857] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/22/2018] [Accepted: 06/15/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Gene L. Bowman
- Nestlé Institute of Health SciencesEPFL CampusLausanneSwitzerland
| | - Loïc Dayon
- Nestlé Institute of Health SciencesEPFL CampusLausanneSwitzerland
| | | | | | - Gwendoline Peyratout
- Old Age PsychiatryDepartment of PsychiatryUniversity Hospital of LausanneLausanneSwitzerland
| | - India C. Severin
- Nestlé Institute of Health SciencesEPFL CampusLausanneSwitzerland
| | - Hugues Henry
- CHUVDepartment of LaboratoriesLausanneSwitzerland
| | - Aikaterini Oikonomidi
- Old Age PsychiatryDepartment of PsychiatryUniversity Hospital of LausanneLausanneSwitzerland
| | | | - Michael Bacher
- Philipps University of MarburgInstitute of ImmunologyMarburgGermany
| | - Julius Popp
- Old Age PsychiatryDepartment of PsychiatryUniversity Hospital of LausanneLausanneSwitzerland
- Geriatric PsychiatryDepartment of Mental Health and PsychiatryUniversity Hospitals of GenevaGenevaSwitzerland
| |
Collapse
|
88
|
Fang X. Impaired tissue barriers as potential therapeutic targets for Parkinson's disease and amyotrophic lateral sclerosis. Metab Brain Dis 2018; 33:1031-1043. [PMID: 29681010 DOI: 10.1007/s11011-018-0239-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier and the intestinal barrier show signs of disruption in patients with idiopathic Parkinson's disease (PD) and animal models of nigrostriatal degeneration, and likewise in amyotrophic lateral sclerosis (ALS) models. A substantial body of evidence shows that defects in epithelial membrane barriers, both in the gut and within the cerebral vasculature, can result in increased vulnerability of tissues to external factors potentially participating in the pathogenesis of PD and ALS. As such, restoration of tissue barriers may prove to be a novel therapeutic target in neurodegenerative disease. In this review, we focus on the potential of new intervention strategies for rescuing and maintaining barrier functions in PD and ALS.
Collapse
Affiliation(s)
- Xin Fang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
89
|
Abstract
Few studies have investigated the role of inflammation in Lewy body dementia (LBD) and variable results have been found. We systematically reviewed the literature for evidence of systemic inflammatory changes in dementia with Lewy bodies and Parkinson disease dementia. Owing to the low number of studies we also included Parkinson disease. Key terms were used to search the relevant databases. Titles and abstracts were screened and potentially relevant articles were reviewed in full. References of included studies and relevant reviews were searched. The database search returned 2166 results, 46 of which were finally included in the systematic review. These studies showed a general increase in inflammatory markers in the peripheral blood, most notably interleukin-1β (IL-1β), tumor necrosis factor-α, IL-6, and IL-10. Studies examining cerebrospinal fluid found IL-1β, IL-6, and transforming growth factor-β1 to be particularly increased, and interferon-γ decreased. C-reactive protein levels were increased, particularly in Parkinson disease dementia. These results provide evidence that LBD is associated with an increased inflammatory response. Furthermore, there may be a stronger general inflammatory response in LBD than in Parkinson disease, while complex changes occur in the individual cytokines.
Collapse
|
90
|
Choi JS, Bae WY, Nam S, Jeong JW. New Targets for Parkinson's Disease: Adhesion G Protein-Coupled Receptor B1 is Downregulated by AMP-Activated Protein Kinase Activation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 22:493-501. [PMID: 30004846 DOI: 10.1089/omi.2018.0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
While progressive dopaminergic neurodegeneration is responsible for the cardinal motor defects in Parkinson's disease (PD), new diagnostics and therapeutic targets are necessary to effectively address this major global health burden. We evaluated whether the adhesion G protein-coupled receptor B1 (ADGRB1, formerly BAI1, brain-specific angiogenesis inhibitor 1) might contribute to dopaminergic neuronal loss. We used bioinformatic analyses, as well as in vitro and in vivo PD models. We report in this study that ADGRB1 is decreased in PD and that the ADGRB1 level is specifically decreased in dopaminergic neurons in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. In primary mouse mesencephalic neurons and human neuroblastoma cell lines, 1-methyl-4-phenylpyridinium (MPP+), a toxic metabolite of MPTP, suppressed the expression of ADGRB1. Moreover, we applied a network generation tool, Ingenuity Pathway Analysis®, with the transcriptomics dataset to extend the upstream regulatory pathway of ADGRB1 expression. AMP-activated protein kinase (AMPK) was predicted as a regulator, and consequently, 5-aminoimidazole-4-carboxamide ribonucleotide, a specific activator of AMPK, reduced the ADGRB1 protein level. Finally, ADGRB1 overexpression decreased nuclear condensation induced by MPP+ treatment. Taken together, we observed that decreased ADGRB1 by activation of AMPK induced neuronal cell death in MPTP/MPP+-mediated PD models, suggesting that ADGRB1 might potentially play a survival role in the neurodegenerative pathway of PD. These data offer new insights into dopaminergic cell death with therapeutic implications for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jae-Sun Choi
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Woom-Yee Bae
- 2 Department of Biomedical Science, Graduate School, Kyung Hee University , Seoul, Republic of Korea
| | - Seungyoon Nam
- 3 Department of Genome Medicine and Science, College of Medicine, Gachon University , Incheon, Republic of Korea.,4 Department of Life Sciences, Gachon University , Seongnam, Republic of Korea.,5 Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center , Incheon, Republic of Korea
| | - Joo-Won Jeong
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University , Seoul, Republic of Korea.,2 Department of Biomedical Science, Graduate School, Kyung Hee University , Seoul, Republic of Korea
| |
Collapse
|
91
|
Zhang J, Zhang Z, Zhang W, Li X, Wu T, Li T, Cai M, Yu Z, Xiang J, Cai D. Jia-Jian-Di-Huang-Yin-Zi decoction exerts neuroprotective effects on dopaminergic neurons and their microenvironment. Sci Rep 2018; 8:9886. [PMID: 29959371 PMCID: PMC6026152 DOI: 10.1038/s41598-018-27852-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/07/2018] [Indexed: 11/09/2022] Open
Abstract
As a classical prescription of Traditional Chinese medicine, the Jia-Jian-Di-Huang-Yin-Zi (JJDHYZ) decoction has long been used to treat movement disorders. The present study evaluated the effects of JJDHYZ on dopaminergic (DA) neurons and their survival-enhancing microenvironment as well as the possible mechanisms involved using a mouse model of Parkinson's disease. In MPTP-lesioned mice, a high dosage of JJDHYZ (34 g/kg/day) attenuated the loss of DA neurons, reversed the dopamine depletion, and improved the expression of glial-derived neurotrophic factor (GDNF) compared to the untreated model group. JJDHYZ also protected the ultrastructure of the blood-brain barrier (BBB) and tight junction proteins by inhibiting the activation of microglia and astrocytes besides the increase in three types of matrix metalloproteinases in the substantia nigra. In conclusion, the JJDHYZ-high dosage (JJDHYZ-H) group exhibited the neuroprotection of DA neurons, and the underlying mechanism may be related to the survival-enhancing microenvironment of the DA neurons.
Collapse
Affiliation(s)
- Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhennian Zhang
- Department of Neurology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing, 210000, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tingting Li
- Department of Neurology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhonghai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
92
|
Alkadhi KA. Exercise as a Positive Modulator of Brain Function. Mol Neurobiol 2018; 55:3112-3130. [PMID: 28466271 DOI: 10.1007/s12035-017-0516-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 04/04/2017] [Indexed: 12/24/2022]
Abstract
Various forms of exercise have been shown to prevent, restore, or ameliorate a variety of brain disorders including dementias, Parkinson's disease, chronic stress, thyroid disorders, and sleep deprivation, some of which are discussed here. In this review, the effects on brain function of various forms of exercise and exercise mimetics in humans and animal experiments are compared and discussed. Possible mechanisms of the beneficial effects of exercise including the role of neurotrophic factors and others are also discussed.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
93
|
Houser MC, Chang J, Factor SA, Molho ES, Zabetian CP, Hill-Burns EM, Payami H, Hertzberg VS, Tansey MG. Stool Immune Profiles Evince Gastrointestinal Inflammation in Parkinson's Disease. Mov Disord 2018; 33:793-804. [PMID: 29572994 DOI: 10.1002/mds.27326] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/17/2017] [Accepted: 01/11/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Gastrointestinal symptoms are common in Parkinson's disease and frequently precede the development of motor impairments. Intestinal inflammation has been proposed as a driver of disease pathology, and evaluation of inflammatory mediators in stool could possibly identify valuable early-stage biomarkers. We measured immune- and angiogenesis-related proteins in human stool to examine inflammatory profiles associated with Parkinson's disease. METHODS Stool samples and subjects' self-reported metadata were obtained from 156 individuals with Parkinson's disease and 110 without, including spouse and nonhousehold controls. Metadata were probed for disease-associated differences, and levels of 37 immune and angiogenesis factors in stool homogenates were measured by multiplexed immunoassay and compared across experimental groups. RESULTS Parkinson's disease patients reported greater incidence of intestinal disease and digestive problems than controls. Direct comparison of levels of stool analytes in patients and controls revealed elevated vascular endothelial growth factor receptor 1, interleukin-1α, and CXCL8 in patients' stool. Paired comparison of patients and spouses suggested higher levels of multiple factors in patients, but this was complicated by sex differences. Sex, body mass index, a history of smoking, and use of probiotics were found to strongly influence levels of stool analytes. Multivariate analysis accounting for these and other potential confounders confirmed elevated levels of interleukin-1α and CXCL8 and also revealed increased interleukin-1β and C-reactive protein in stool in Parkinson's disease. These differences were not dependent on subject age or disease duration. CONCLUSIONS Levels of stool immune factors indicate that intestinal inflammation is present in patients with Parkinson's disease. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Madelyn C Houser
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jianjun Chang
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stewart A Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eric S Molho
- Department of Neurology, Albany Medical College, Albany, New York, USA
| | - Cyrus P Zabetian
- Veterans Affairs Puget Sound Health Care System and Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Erin M Hill-Burns
- Department of Neurology, University of Alabama at Birmingham, Birminham, Alabama, USA
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birminham, Alabama, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Vicki S Hertzberg
- Center for Nursing Data Science, Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, Georgia, USA
| | - Malú G Tansey
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
94
|
Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 2018; 14:133-150. [PMID: 29377008 PMCID: PMC5829048 DOI: 10.1038/nrneurol.2017.188] [Citation(s) in RCA: 1945] [Impact Index Per Article: 277.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) is a continuous endothelial membrane within brain microvessels that has sealed cell-to-cell contacts and is sheathed by mural vascular cells and perivascular astrocyte end-feet. The BBB protects neurons from factors present in the systemic circulation and maintains the highly regulated CNS internal milieu, which is required for proper synaptic and neuronal functioning. BBB disruption allows influx into the brain of neurotoxic blood-derived debris, cells and microbial pathogens and is associated with inflammatory and immune responses, which can initiate multiple pathways of neurodegeneration. This Review discusses neuroimaging studies in the living human brain and post-mortem tissue as well as biomarker studies demonstrating BBB breakdown in Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, multiple sclerosis, HIV-1-associated dementia and chronic traumatic encephalopathy. The pathogenic mechanisms by which BBB breakdown leads to neuronal injury, synaptic dysfunction, loss of neuronal connectivity and neurodegeneration are described. The importance of a healthy BBB for therapeutic drug delivery and the adverse effects of disease-initiated, pathological BBB breakdown in relation to brain delivery of neuropharmaceuticals are briefly discussed. Finally, future directions, gaps in the field and opportunities to control the course of neurological diseases by targeting the BBB are presented.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo Street, Los Angeles, California 90089, USA
| |
Collapse
|
95
|
Zou J, Chen Z, Liang C, Fu Y, Wei X, Lu J, Pan M, Guo Y, Liao X, Xie H, Wu D, Li M, Liang L, Wang P, Wang Q. Trefoil Factor 3, Cholinesterase and Homocysteine: Potential Predictors for Parkinson's Disease Dementia and Vascular Parkinsonism Dementia in Advanced Stage. Aging Dis 2018; 9:51-65. [PMID: 29392081 PMCID: PMC5772858 DOI: 10.14336/ad.2017.0416] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/16/2017] [Indexed: 01/23/2023] Open
Abstract
Trefoil factor 3 (TFF3), cholinesterase activity (ChE activity) and homocysteine (Hcy) play critical roles in modulating recognition, learning and memory in neurodegenerative diseases, such as Parkinson's disease dementia (PDD) and vascular parkinsonism with dementia (VPD). However, whether they can be used as reliable predictors to evaluate the severity and progression of PDD and VPD remains largely unknown. METHODS We performed a cross-sectional study that included 92 patients with PDD, 82 patients with VPD and 80 healthy controls. Serum levels of TFF3, ChE activity and Hcy were measured. Several scales were used to rate the severity of PDD and VPD. Receivers operating characteristic (ROC) curves were applied to map the diagnostic accuracy of PDD and VPD patients compared to healthy subjects. RESULTS Compared with healthy subjects, the serum levels of TFF3 and ChE activity were lower, while Hcy was higher in the PDD and VPD patients. These findings were especially prominent in male patients. The three biomarkers displayed differences between PDD and VPD sub-groups based on genders and UPDRS (III) scores' distribution. Interestingly, these increased serum Hcy levels were significantly and inversely correlated with decreased TFF3/ChE activity levels. There were significant correlations between TFF3/ChE activity/Hcy levels and PDD/VPD severities, including motor dysfunction, declining cognition and mood/gastrointestinal symptoms. Additionally, ROC curves for the combination of TFF3, ChE activity and Hcy showed potential diagnostic value in discriminating PDD and VPD patients from healthy controls. CONCLUSIONS Our findings suggest that serum TFF3, ChE activity and Hcy levels may underlie the pathophysiological mechanisms of PDD and VPD. As the race to find biomarkers or predictors for these diseases intensifies, a better understanding of the roles of TFF3, ChE activity and Hcy may yield insights into the pathogenesis of PDD and VPD.
Collapse
Affiliation(s)
| | | | - Caiqian Liang
- 2Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, China
| | - Yongmei Fu
- 2Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, China
| | | | - Jianjun Lu
- 3Department of Neurology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Mengqiu Pan
- 3Department of Neurology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Yue Guo
- 4Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Xinxue Liao
- 4Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Huifang Xie
- 5Department of Neurology, Zhujiang Hospital, Southern Medical University, China
| | - Duobin Wu
- 5Department of Neurology, Zhujiang Hospital, Southern Medical University, China
| | - Min Li
- 6School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Lihui Liang
- 7Department of Geriatric Medicine, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Penghua Wang
- 8Department of Microbiology & Immunology, School of Medicine, New York Medical College, NY 10595, USA
| | | |
Collapse
|
96
|
Shim JW, Madsen JR. VEGF Signaling in Neurological Disorders. Int J Mol Sci 2018; 19:ijms19010275. [PMID: 29342116 PMCID: PMC5796221 DOI: 10.3390/ijms19010275] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/06/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent growth factor playing diverse roles in vasculogenesis and angiogenesis. In the brain, VEGF mediates angiogenesis, neural migration and neuroprotection. As a permeability factor, excessive VEGF disrupts intracellular barriers, increases leakage of the choroid plexus endothelia, evokes edema, and activates the inflammatory pathway. Recently, we discovered that a heparin binding epidermal growth factor like growth factor (HB-EGF)—a class of EGF receptor (EGFR) family ligands—contributes to the development of hydrocephalus with subarachnoid hemorrhage through activation of VEGF signaling. The objective of this review is to entail a recent update on causes of death due to neurological disorders involving cerebrovascular and age-related neurological conditions and to understand the mechanism by which angiogenesis-dependent pathological events can be treated with VEGF antagonisms. The Global Burden of Disease study indicates that cancer and cardiovascular disease including ischemic and hemorrhagic stroke are two leading causes of death worldwide. The literature suggests that VEGF signaling in ischemic brains highlights the importance of concentration, timing, and alternate route of modulating VEGF signaling pathway. Molecular targets distinguishing two distinct pathways of VEGF signaling may provide novel therapies for the treatment of neurological disorders and for maintaining lower mortality due to these conditions.
Collapse
Affiliation(s)
- Joon W Shim
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
97
|
Jourdain VA, Schindlbeck KA, Tang CC, Niethammer M, Choi YY, Markowitz D, Nazem A, Nardi D, Carras N, Feigin A, Ma Y, Peng S, Dhawan V, Eidelberg D. Increased putamen hypercapnic vasoreactivity in levodopa-induced dyskinesia. JCI Insight 2017; 2:96411. [PMID: 29046477 DOI: 10.1172/jci.insight.96411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/07/2017] [Indexed: 12/29/2022] Open
Abstract
In a rodent model of Parkinson's disease (PD), levodopa-induced involuntary movements have been linked to striatal angiogenesis - a process that is difficult to document in living human subjects. Angiogenesis can be accompanied by localized increases in cerebral blood flow (CBF) responses to hypercapnia. We therefore explored the possibility that, in the absence of levodopa, local hypercapnic CBF responses are abnormally increased in PD patients with levodopa-induced dyskinesias (LID) but not in their nondyskinetic (NLID) counterparts. We used H215O PET to scan 24 unmedicated PD subjects (12 LID and 12 NLID) and 12 matched healthy subjects in the rest state under normocapnic and hypercapnic conditions. Hypercapnic CBF responses were compared to corresponding levodopa responses from the same subjects. Group differences in hypercapnic vasoreactivity were significant only in the posterior putamen, with greater CBF responses in LID subjects compared with the other subjects. Hypercapnic and levodopa-mediated CBF responses measured in this region exhibited distinct associations with disease severity: the former correlated with off-state motor disability ratings but not symptom duration, whereas the latter correlated with symptom duration but not motor disability. These are the first in vivo human findings linking LID to microvascular changes in the basal ganglia.
Collapse
Affiliation(s)
- Vincent A Jourdain
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Katharina A Schindlbeck
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Chris C Tang
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Martin Niethammer
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| | - Yoon Young Choi
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | | | - Amir Nazem
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Dominic Nardi
- Department of Anesthesiology, Northwell Health, Manhasset, New York, USA
| | - Nicholas Carras
- Department of Anesthesiology, Northwell Health, Manhasset, New York, USA
| | - Andrew Feigin
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| | - Yilong Ma
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Shichun Peng
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Vijay Dhawan
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - David Eidelberg
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| |
Collapse
|
98
|
The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron 2017; 96:17-42. [PMID: 28957666 DOI: 10.1016/j.neuron.2017.07.030] [Citation(s) in RCA: 1507] [Impact Index Per Article: 188.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/20/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
Abstract
The concept of the neurovascular unit (NVU), formalized at the 2001 Stroke Progress Review Group meeting of the National Institute of Neurological Disorders and Stroke, emphasizes the intimate relationship between the brain and its vessels. Since then, the NVU has attracted the interest of the neuroscience community, resulting in considerable advances in the field. Here the current state of knowledge of the NVU will be assessed, focusing on one of its most vital roles: the coupling between neural activity and blood flow. The evidence supports a conceptual shift in the mechanisms of neurovascular coupling, from a unidimensional process involving neuronal-astrocytic signaling to local blood vessels to a multidimensional one in which mediators released from multiple cells engage distinct signaling pathways and effector systems across the entire cerebrovascular network in a highly orchestrated manner. The recently appreciated NVU dysfunction in neurodegenerative diseases, although still poorly understood, supports emerging concepts that maintaining neurovascular health promotes brain health.
Collapse
|
99
|
Al-Bachari S, Vidyasagar R, Emsley HC, Parkes LM. Structural and physiological neurovascular changes in idiopathic Parkinson's disease and its clinical phenotypes. J Cereb Blood Flow Metab 2017; 37:3409-3421. [PMID: 28112022 PMCID: PMC5624390 DOI: 10.1177/0271678x16688919] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neurovascular changes are likely to interact importantly with the neurodegenerative process in idiopathic Parkinson's disease (IPD). Markers of neurovascular status (NVS) include white matter lesion (WML) burden and arterial spin labelling (ASL) measurements of cerebral blood flow (CBF) and arterial arrival time (AAT). We investigated NVS in IPD, including an analysis of IPD clinical phenotypes, by comparison with two control groups, one with a history of clinical cerebrovascular disease (CVD) (control positive, CP) and one without CVD (control negative, CN). Fifty-one patients with IPD (mean age 69.0 ± 7.7 years) (21 tremor dominant (TD), 24 postural instability and gait disorder (PIGD) and six intermediates), 18 CP (mean age 70.1 ± 8.0 years) and 34 CN subjects (mean age 67.4 ± 7.6 years) completed a 3T MRI scan protocol including T2-weighted fluid-attenuated inversion recovery (FLAIR) and ASL. IPD patients showed diffuse regions of significantly prolonged AAT, small regions of lower CBF and greater WML burden by comparison with CN subjects. TD patients showed lower WML volume by comparison with PIGD patients. These imaging data thus show altered NVS in IPD, with some evidence for IPD phenotype-specific differences.
Collapse
Affiliation(s)
- Sarah Al-Bachari
- 1 Department of Neurology, Salford Royal NHS Foundation Trust, Salford, UK.,2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,3 Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Rishma Vidyasagar
- 2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,4 Anatomy and Neuroscience Department, University of Melbourne, Melbourne, Australia.,5 Florey Institute of Neuroscience and Mental Health, Heidelberg, Melbourne, Australia
| | - Hedley Ca Emsley
- 6 Department of Neurology, Royal Preston Hospital, Preston, UK.,7 Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Laura M Parkes
- 2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,8 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
100
|
Zou J, Chen Z, Wei X, Chen Z, Fu Y, Yang X, Chen D, Wang R, Jenner P, Lu JH, Li M, Zhang Z, Tang B, Jin K, Wang Q. Cystatin C as a potential therapeutic mediator against Parkinson's disease via VEGF-induced angiogenesis and enhanced neuronal autophagy in neurovascular units. Cell Death Dis 2017; 8:e2854. [PMID: 28569795 PMCID: PMC5520899 DOI: 10.1038/cddis.2017.240] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023]
Abstract
Cystatin C (CYS C, Cst3) is an endogenous cysteine protease inhibitor that plays neuroprotective roles in neurodegenerative diseases. We aimed to explore the association of CYS C with Parkinson’s disease (PD) models and investigate its involvement in the role of neurovascular units (NVUs) in PD neuro-pathogenesis. We used A53T α-synuclein (SNCA) transgenic mice and 6-hydroxydopamine-lesioned DAergic PC12 cells as experimental PD models to investigate the mechanisms behind this association. The injections of CYS C were administered to the right substantia nigra (SN) of A53T SNCA transgenic mice to measure the effects of CYS C in transgenic A53T SNCA mice. To explore the angiogenesis in vivo and in vitro, we used the chick embryo chorioallantoic membrane (CAM) assay and tube formation (TF) assay. We found that CYS C has a neuroprotective effect in this in vivo PD model. We observed increased VEGF, NURR1 and autophagy markers LC3B and decreased SNCA and apoptosis marker cleaved CASP3 in different brain regions of CYS C-treated A53T SNCA transgenic mice. In vitro, we observed that CYS C-induced VEGF, a secreted protein, attenuated 6-OHDA-lesioned DAergic PC12 cell degeneration by regulating p-PKC-α/p-ERK1/2-Nurr1 signaling and inducing autophagy. VEGF-mediated angiogenesis was markedly enhanced in the conditioned media of 6-OHDA-lesioned PC12 cells with CYS C-overexpression, whereas blockage of autophagy in CYS C-overexpressing PC12 cells significantly downregulated VEGF expression and the associated angiogenesis. Our data indicate that CYS C displays dual neuronal–vascular functions, promoting PC12 cell survival and angiogenesis via regulating the level of secreted VEGF in NVUs. Our study provides evidence that may aid in the development of an alternative approach for the treatment of PD through modulation of CYS C-mediated neuronal-vascular pathways.
Collapse
Affiliation(s)
- Jing Zou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhaoyu Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaobo Wei
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhigang Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Yongmei Fu
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoyan Yang
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Dan Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Rui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Faculty of Health Sciences and Medicine, King's College, London SE1 1UL, UK
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhuohua Zhang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Beisha Tang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qing Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|