1
|
Yadav S, Bukke SPN, Prajapati S, Singh AP, Chettupalli AK, Nicholas B. Nanobiosensors in neurodegenerative disease diagnosis: A promising pathway for early detection. Digit Health 2025; 11:20552076251342457. [PMID: 40376568 PMCID: PMC12078979 DOI: 10.1177/20552076251342457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/28/2025] [Indexed: 05/18/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's and Parkinson's, are characterized by progressive neuronal loss, leading to cognitive and motor impairments. Early diagnosis remains a challenge due to the slow progression of symptoms and the limitations of current diagnostic methods. Nanobiosensors, leveraging the high sensitivity and specificity of nanotechnology, offer a promising, noninvasive, and cost-effective approach for detecting disease biomarkers at ultra-low concentrations. This review highlights recent advancements in nanobiosensor technology, including the integration of gold nanoparticles, quantum dots, and carbon nanotubes, which have significantly enhanced biomarker detection precision. Furthermore, it examines the advantages of nanobiosensors over traditional diagnostic techniques, such as improved sensitivity, rapid detection, and minimal invasiveness. The potential of these innovative sensors to revolutionize early disease detection and improve patient outcomes is discussed, along with existing challenges in clinical translation, including stability, reproducibility, and regulatory considerations. Addressing these limitations will be crucial for integrating nanobiosensors into routine clinical practice and advancing personalized medicine for neurodegenerative disorders.
Collapse
Affiliation(s)
- Shikha Yadav
- Department of Pharmaceutical Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Sarad Pawar Naik Bukke
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Ishaka-Bushenyi, Uganda
| | | | - Ajay Pal Singh
- School of Pharmacy, Lingaya's Vidyapeeth, Faridabad, Haryana, India
| | - Ananda Kumar Chettupalli
- Department of Pharmaceutical Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Buyinza Nicholas
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Ishaka-Bushenyi, Uganda
| |
Collapse
|
2
|
Kluge A, Schaeffer E, Bunk J, Sommerauer M, Röttgen S, Schulte C, Roeben B, von Thaler AK, Welzel J, Lucius R, Heinzel S, Xiang W, Eschweiler GW, Maetzler W, Suenkel U, Berg D. Detecting Misfolded α-Synuclein in Blood Years before the Diagnosis of Parkinson's Disease. Mov Disord 2024; 39:1289-1299. [PMID: 38651526 DOI: 10.1002/mds.29766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Identifying individuals with Parkinson's disease (PD) already in the prodromal phase of the disease has become a priority objective for opening a window for early disease-modifying therapies. OBJECTIVE The aim was to evaluate a blood-based α-synuclein seed amplification assay (α-syn SAA) as a novel biomarker for diagnosing PD in the prodromal phase. METHODS In the TREND study (University of Tuebingen) biennial blood samples of n = 1201 individuals with/without increased risk for PD were taken prospectively over 4 to 10 years. We retrospectively analyzed blood samples of 12 participants later diagnosed with PD during the study to detect and amplify pathological α-syn conformers derived from neuronal extracellular vesicles using (1) immunoblot analyses with an antibody against these conformers and (2) an α-syn-SAA. Additionally, blood samples of n = 13 healthy individuals from the TREND cohort and n = 20 individuals with isolated rapid eye movement sleep behavior disorder (iRBD) from the University Hospital Cologne were analyzed. RESULTS All individuals with PD showed positive immunoblots and a positive α-syn SAA at the time of diagnosis. Moreover, all PD patients showed a positive α-syn SAA 1 to 10 years before clinical diagnosis. In the iRBD cohort, 30% showed a positive α-syn SAA. All healthy controls had a negative SAA. CONCLUSIONS We here demonstrate the possibility to detect and amplify pathological α-syn conformers in peripheral blood up to 10 years before the clinical diagnosis of PD in individuals with and without iRBD. The findings of this study indicate that this blood-based α-syn SAA assay has the potential to serve as a diagnostic biomarker for prodromal PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Josina Bunk
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Michael Sommerauer
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany
| | - Sinah Röttgen
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Benjamin Roeben
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Anna-Katharina von Thaler
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Julius Welzel
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Ralph Lucius
- Institute of Anatomy, Kiel University, Kiel, Germany
| | - Sebastian Heinzel
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Gerhard W Eschweiler
- Geriatric Center, University Hospital Tübingen, Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Ulrike Suenkel
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Partner Site Tübingen, Tübingen, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
3
|
Luo X, Liu Y, Balck A, Klein C, Fleming RMT. Identification of metabolites reproducibly associated with Parkinson's Disease via meta-analysis and computational modelling. NPJ Parkinsons Dis 2024; 10:126. [PMID: 38951523 PMCID: PMC11217404 DOI: 10.1038/s41531-024-00732-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/30/2024] [Indexed: 07/03/2024] Open
Abstract
Many studies have reported metabolomic analysis of different bio-specimens from Parkinson's disease (PD) patients. However, inconsistencies in reported metabolite concentration changes make it difficult to draw conclusions as to the role of metabolism in the occurrence or development of Parkinson's disease. We reviewed the literature on metabolomic analysis of PD patients. From 74 studies that passed quality control metrics, 928 metabolites were identified with significant changes in PD patients, but only 190 were replicated with the same changes in more than one study. Of these metabolites, 60 exclusively increased, such as 3-methoxytyrosine and glycine, 54 exclusively decreased, such as pantothenic acid and caffeine, and 76 inconsistently changed in concentration in PD versus control subjects, such as ornithine and tyrosine. A genome-scale metabolic model of PD and corresponding metabolic map linking most of the replicated metabolites enabled a better understanding of the dysfunctional pathways of PD and the prediction of additional potential metabolic markers from pathways with consistent metabolite changes to target in future studies.
Collapse
Affiliation(s)
- Xi Luo
- School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Yanjun Liu
- School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Alexander Balck
- Institute of Neurogenetics and Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Christine Klein
- Institute of Neurogenetics and Department of Neurology, University of Luebeck and University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Ronan M T Fleming
- School of Medicine, University of Galway, University Rd, Galway, Ireland.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands.
| |
Collapse
|
4
|
Wang Q, Wang H, Zhao X, Han C, Liu C, Li Z, Du T, Sui Y, Zhang X, Zhang J, Xiao Y, Cai G, Meng F. Transcriptome sequencing of circular RNA reveals the involvement of hsa-SCMH1_0001 in the pathogenesis of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14435. [PMID: 37664885 PMCID: PMC10916443 DOI: 10.1111/cns.14435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/01/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease. Exosomes are endosome-derived extracellular vesicles that can take part in intercellular communication. Circular RNAs (circRNAs) are noncoding RNAs characterized by covalently closed-loop structures, which perform a crucial function in many diseases. AIM To clarify the expression and function of exosomal circRNSs of PD patients and look for circRNAs that might be related to the pathogenesis of PD. MATERIALS AND METHODS We examined circRNA and mRNA expression profiles in peripheral exosomes from PD patients (n = 23) and healthy controls (n = 15) using next-generation sequencing (NGS) technology, functional annotation, and quantitative polymerase chain reaction. Correlation analysis was performed between the expression levels of the circRNAs and the clinical characteristics of PD patients. The binding miRNAs and target genes were predicted using TargetScanHuman, miRDB, and miRTarBase. The predicted target genes were compared with the differentially expressed mRNAs in sequencing results. RESULTS According to the NGS, 62 upregulated and 37 downregulated circRNAs in the PD group were screened out. Correlation analysis revealed that hsa-SCMH1_0001 has strong clinical relevance. We identified 17 potential binding miRNAs of hsa-SCMH1_0001 with 149 potential target genes. ARID1A and C1orf115 belong to the intersection of the predicted target genes and the differentially expressed mRNAs obtained by sequencing. CONCLUSION This study suggested that hsa-SCMH1_0001 and its target genes ARID1A and C1orf115 are downregulated in PD patients and may be involved in the occurrence of PD.
Collapse
Affiliation(s)
- Qiao Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
- National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijing HospitalBeijingChina
| | - Huizhi Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Xuemin Zhao
- Department of Neurophysiology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Chunlei Han
- Beijing Key Laboratory of NeurostimulationBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chong Liu
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Zhibao Li
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Tingting Du
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Yunpeng Sui
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Xin Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
| | - Jianguo Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yilei Xiao
- Department of NeurosurgeryLiaocheng People's HospitalLiaochengChina
| | - Guoen Cai
- Department of NeurologyFujian Medical University Union HospitalFuzhouChina
- Fujian Key Laboratory of Molecular Neurology, Institute of Clinical Neurology, Institute of NeuroscienceFujian Medical UniversityFuzhouChina
| | - Fangang Meng
- Department of Functional Neurosurgery, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of NeurostimulationBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| |
Collapse
|
5
|
Vijayakumari AA, Fernandez HH, Walter BL. MRI-based multivariate gray matter volumetric distance for predicting motor symptom progression in Parkinson's disease. Sci Rep 2023; 13:17704. [PMID: 37848592 PMCID: PMC10582255 DOI: 10.1038/s41598-023-44322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 10/19/2023] Open
Abstract
While Parkinson's disease (PD)-related neurodegeneration is associated with structural changes in the brain, conventional magnetic resonance imaging (MRI) has proven less effective for clinical diagnosis due to its inability to reliably identify subtle changes early in the disease course. In this study, we aimed to develop a structural MRI-based biomarker to predict the rate of progression of motor symptoms in the early stages of PD. The study included 88 patients with PD and 120 healthy controls from the Parkinson's Progression Markers Initiative database; MRI at baseline and motor symptom scores assessed using the MDS-UPDRS-III at two time points (baseline and 48 months) were selected. Group-level volumetric analyses revealed that the volumetric reductions in the left striatum were associated with the decline in motor functioning. Then, we developed a patient-specific multivariate gray matter volumetric distance and demonstrated that it could significantly predict changes in motor symptom scores (P < 0.05). Further, we classified patients as relatively slower and faster progressors with 89% accuracy using a support vector machine classifier. Thus, we identified a promising structural MRI-based biomarker for predicting the rate of progression of motor symptoms and classifying patients based on motor symptom severity.
Collapse
Affiliation(s)
- Anupa A Vijayakumari
- Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, Mail Code: S20, Cleveland, OH, 44195, USA
| | - Hubert H Fernandez
- Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, Mail Code: S20, Cleveland, OH, 44195, USA
| | - Benjamin L Walter
- Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Avenue, Mail Code: S20, Cleveland, OH, 44195, USA.
| |
Collapse
|
6
|
Dutta S, Hornung S, Taha HB, Bitan G. Biomarkers for parkinsonian disorders in CNS-originating EVs: promise and challenges. Acta Neuropathol 2023; 145:515-540. [PMID: 37012443 PMCID: PMC10071251 DOI: 10.1007/s00401-023-02557-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023]
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and oncosomes, are nano-sized particles enclosed by a lipid bilayer. EVs are released by virtually all eukaryotic cells and have been shown to contribute to intercellular communication by transporting proteins, lipids, and nucleic acids. In the context of neurodegenerative diseases, EVs may carry toxic, misfolded forms of amyloidogenic proteins and facilitate their spread to recipient cells in the central nervous system (CNS). CNS-originating EVs can cross the blood-brain barrier into the bloodstream and may be found in other body fluids, including saliva, tears, and urine. EVs originating in the CNS represent an attractive source of biomarkers for neurodegenerative diseases, because they contain cell- and cell state-specific biological materials. In recent years, multiple papers have reported the use of this strategy for identification and quantitation of biomarkers for neurodegenerative diseases, including Parkinson's disease and atypical parkinsonian disorders. However, certain technical issues have yet to be standardized, such as the best surface markers for isolation of cell type-specific EVs and validating the cellular origin of the EVs. Here, we review recent research using CNS-originating EVs for biomarker studies, primarily in parkinsonian disorders, highlight technical challenges, and propose strategies for overcoming them.
Collapse
Affiliation(s)
- Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, India
| | - Simon Hornung
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hash Brown Taha
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA.
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Painous C, Compta Y. Sacades, pupils and blink tracking: More than meets the eye in the Parkinson's disease cognitive spectrum? Parkinsonism Relat Disord 2023; 110:105363. [PMID: 36966052 DOI: 10.1016/j.parkreldis.2023.105363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Affiliation(s)
- Celia Painous
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic I Universitari de Barcelona, IDIBAPS, CIBERNED (CB06/05/0018-ISCIII), ERN- RND, Institut Clínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic I Universitari de Barcelona, IDIBAPS, CIBERNED (CB06/05/0018-ISCIII), ERN- RND, Institut Clínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
8
|
Sharma V, Nikolajeff F, Kumar S. Employing nanoparticle tracking analysis of salivary neuronal exosomes for early detection of neurodegenerative diseases. Transl Neurodegener 2023; 12:7. [PMID: 36747288 PMCID: PMC9903484 DOI: 10.1186/s40035-023-00339-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative diseases are a set of progressive and currently incurable diseases that are primarily caused by neuron degeneration. Neurodegenerative diseases often lead to cognitive impairment and dyskinesias. It is now well recognized that molecular events precede the onset of clinical symptoms by years. Over the past decade, intensive research attempts have been aimed at the early diagnosis of these diseases. Recently, exosomes have been shown to play a pivotal role in the occurrence and progression of many diseases including cancer and neurodegenerative diseases. Additionally, because exosomes can cross the blood-brain barrier, they may serve as a diagnostic tool for neural dysfunction. In this review, we detail the mechanisms and current challenges of these diseases, briefly review the role of exosomes in the progression of neurodegenerative diseases, and propose a novel strategy based on salivary neuronal exosomes and nanoparticle tracking analysis that could be employed for screening the early onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Vaibhav Sharma
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden.
| | - Fredrik Nikolajeff
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden
| | - Saroj Kumar
- Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden.
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
9
|
Sheng ZH, Ma LZ, Liu JY, Ou YN, Zhao B, Ma YH, Tan L. Cerebrospinal fluid neurofilament dynamic profiles predict cognitive progression in individuals with de novo Parkinson's disease. Front Aging Neurosci 2022; 14:1061096. [PMID: 36589544 PMCID: PMC9802677 DOI: 10.3389/fnagi.2022.1061096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Background Neurofilament light chain protein (NfL) in cerebrospinal fluid (CSF) reflects the severity of neurodegeneration, with its altered concentrations discovered in Parkinson's disease (PD) and Parkinson's disease dementia (PD-D). Objective To determine whether CSF NfL, a promising biomarker of neuronal/axonal damage, can be used to monitor cognitive progression in de novo Parkinson's disease and predict future cognitive decline. Methods A total of 259 people were recruited in this study, including 85 healthy controls (HC) and 174 neonatal PD patients from the Parkinson's Progression Markers Initiative (PPMI). Multiple linear regression and linear mixed effects models were used to examine the associations of baseline/longitudinal CSF NfL with cognitive decline and other CSF biomarkers. Kaplan-Meier analysis and log-rank test were used to compare the cumulative probability risk of cognition progression during the follow-up. Multivariate cox regression was used to detect cognitive progression in de novo PD. Results We found PD patients with mild cognitive impairment (PD-MCI) was higher than with normal cognition (PD-NC) in terms of CSF NfL baseline levels (p = 0.003) and longitudinal increase rate (p = 0.034). Both baseline CSF NfL and its rate of change predicted measurable cognitive decline in de novo PD (MoCA, β = -0.010, p = 0.011; β = -0.0002, p < 0.001, respectively). The predictive effects in de novo PD patients aged >65, male, ill-educated (<13 years) and without carrying Apolipoprotein E ε4 (APOE ε4) seemed to be more obvious and reflected in more domains investigated. We also observed that CSF NfL levels predicted progression in de novo PD patients with different cognitive diagnosis and amyloid status. After an average follow-up of 6.66 ± 2.54 years, higher concentration above the median of baseline CSF NfL was associated with a future high risk of PD with dementia (adjusted HR 2.82, 95% CI: 1.11-7.20, p = 0.030). Conclusion Our results indicated that CSF NfL is a promising prognostic predictor of PD, and its concentration and dynamics can monitor the severity and progression of cognitive decline in de novo PD patients.
Collapse
|
10
|
Gramotnev DK, Gramotnev G, Gramotnev A, Summers MJ. Path analysis of biomarkers for cognitive decline in early Parkinson’s disease. PLoS One 2022; 17:e0268379. [PMID: 35560326 PMCID: PMC9106174 DOI: 10.1371/journal.pone.0268379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Clinical and biochemical diversity of Parkinson’s disease (PD) and numerous demographic, clinical, and pathological measures influencing cognitive function and its decline in PD create problems with the determination of effects of individual measures on cognition in PD. This is particularly the case where these measures significantly interrelate with each other producing intricate networks of direct and indirect effects on cognition. Here, we use generalized structural equation modelling (GSEM) to identify and characterize significant paths for direct and indirect effects of 14 baseline measures on global cognition in PD at baseline and at 4 years later. We consider 269 drug-naïve participants from the Parkinson’s Progression Marker Initiative database, diagnosed with idiopathic PD and observed for at least 4 years after baseline. Two GSEM networks are derived, highlighting the possibility of at least two different molecular pathways or two different PD sub-types, with either CSF p-tau181 or amyloid beta (1–42) being the primary protein variables potentially driving progression of cognitive decline. The models provide insights into the interrelations between the 14 baseline variables, and determined their total effects on cognition in early PD. High CSF amyloid concentrations (> 500 pg/ml) are associated with nearly full protection against cognitive decline in early PD in the whole range of baseline age between 40 and 80 years, and irrespectively of whether p-tau181 or amyloid beta (1–42) are considered as the primary protein variables. The total effect of depression on cognition is shown to be strongly amplified by PD, but not at the time of diagnosis or at prodromal stages. CSF p-tau181 protein could not be a reliable indicator of cognitive decline because of its significantly heterogeneous effects on cognition. The outcomes will enable better understanding of the roles of the clinical and pathological measures and their mutual effects on cognition in early PD.
Collapse
Affiliation(s)
| | - Galina Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
| | - Alexandra Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
- Sunshine Coast Mind & Neuroscience – Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Mathew J. Summers
- School of Health and Behavioural Science, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
11
|
Kluge A, Bunk J, Schaeffer E, Drobny A, Xiang W, Knacke H, Bub S, Lückstädt W, Arnold P, Lucius R, Berg D, Zunke F. OUP accepted manuscript. Brain 2022; 145:3058-3071. [PMID: 35722765 DOI: 10.1093/brain/awac115] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/12/2022] [Accepted: 03/13/2022] [Indexed: 11/13/2022] Open
Abstract
To date, no reliable clinically applicable biomarker has been established for Parkinson's disease. Our results indicate that a long anticipated blood test for Parkinson's disease may be realized. Following the isolation of neuron-derived extracellular vesicles of Parkinson's disease patients and non-Parkinson's disease individuals, immunoblot analyses were performed to detect extracellular vesicle-derived α-synuclein. Pathological α-synuclein forms derived from neuronal extracellular vesicles could be detected under native conditions and were significantly increased in all individuals with Parkinson's disease and clearly distinguished disease from the non-disease state. By performing an α-synuclein seeding assay these soluble conformers could be amplified and seeding of pathological protein folding was demonstrated. Amplified α-synuclein conformers exhibited β-sheet-rich structures and a fibrillary appearance. Our study demonstrates that the detection of pathological α-synuclein conformers from neuron-derived extracellular vesicles from blood plasma samples has the potential to evolve into a blood-biomarker of Parkinson's disease that is still lacking so far. Moreover, the distribution of seeding-competent α-synuclein within blood exosomes sheds a new light of pathological disease mechanisms in neurodegenerative disorders.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Josina Bunk
- Institute of Biochemistry, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Henrike Knacke
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Simon Bub
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wiebke Lückstädt
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Ralph Lucius
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
12
|
Liguori C, Stefani A, Fernandes M, Cerroni R, Mercuri NB, Pierantozzi M. Biomarkers of Cerebral Glucose Metabolism and Neurodegeneration in Parkinson's Disease: A Cerebrospinal Fluid-Based Study. JOURNAL OF PARKINSON'S DISEASE 2021; 12:537-544. [PMID: 34864690 DOI: 10.3233/jpd-212936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Several biomarkers have been evaluated in Parkinson's disease (PD); cerebrospinal fluid (CSF) levels of lactate may reflect cerebral metabolism function and CSF amyloid-β42 (Aβ42), total tau (t-tau) and phosphorylated tau (p-tau) concentrations may detect an underlying neurodegenerative process. OBJECTIVE CSF levels of lactate, Aβ42, t-tau, and p-tau were measured in patients with mild to moderate PD. CSF levels of dopamine (DA) and its metabolite 3,4-Dihydroxyphenylacetic acid (DOPAC) were also assessed, exploring their relations with the other CSF biomarkers. METHODS 101 drug-naive PD patients and 60 controls were included. Participants underwent clinical assessments and CSF biomarker analysis. Patients were divided into subgroups according to their Hoehn & Yahr stage (PD-1, PD-2, PD-3). RESULTS PD patients showed higher lactate levels (M = 1.91; p = 0.03) and lower Aβ42 (M = 595; p < 0.001) and DA levels (M = 0.32; p = 0.04) than controls (Mlactate = 1.72; MAβ42 = 837; MDA = 0.50), while no significant differences were found in t-tau, p-tau and DOPAC concentrations. Considering the subgroup analysis, PD-3 group had higher lactate (M = 2.12) and t-tau levels (M = 333) than both PD-1 (Mlactate = 1.75, p = 0.006; Mt - tau = 176, p = 0.008) and PD-2 groups (Mlactate = 1.91, p = 0.01; Mt - tau = 176, p = 0.03), as well as the controls (Mlactate = 1.72, p = 0.04; Mt - tau = 205, p = 0.04). PD-2 group showed higher lactate levels than PD-1 group (p = 0.04) and controls (p = 0.03). Finally, CSF lactate levels negatively correlated with DA (r = -0.42) and positively with t-tau CSF levels (r = 0.33). CONCLUSION This CSF-based study shows that lactate levels in PD correlated with both clinical disease progression and neurodegeneration biomarkers, such as tau proteins and DA. Further studies should explore the clinical potential of measuring CSF biomarkers for better understanding the role of brain energy metabolism in PD, for research and therapeutic options.
Collapse
Affiliation(s)
- Claudio Liguori
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,Sleep Medicine Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,UOSD Parkinson's Disease Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy
| | - Alessandro Stefani
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,UOSD Parkinson's Disease Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy
| | - Mariana Fernandes
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy
| | - Rocco Cerroni
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,UOSD Parkinson's Disease Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | - Mariangela Pierantozzi
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Italy.,UOSD Parkinson's Disease Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Italy
| |
Collapse
|
13
|
Li X, Fan X, Yang H, Liu Y. Review of Metabolomics-Based Biomarker Research for Parkinson's Disease. Mol Neurobiol 2021; 59:1041-1057. [PMID: 34826053 DOI: 10.1007/s12035-021-02657-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/17/2021] [Indexed: 01/12/2023]
Abstract
Parkinson's disease (PD), as the second most common neurodegenerative disease, is seriously affecting the life quality of the elderly. However, there is still a lack of efficient medical methods to diagnosis PD before apparent symptoms occur. In recent years, clinical biomarkers including genetic, imaging, and tissue markers have exhibited remarkable benefits in assisting PD diagnoses. Due to the advantages of high-throughput detection of metabolites and almost non-invasive sample collection, metabolomics research of PD is widely used for diagnostic biomarker discovery. However, there are also a few shortages for those identified biomarkers, such as the scarcity of verifications regarding the sensitivity and specificity. Thus, reviewing the research progress of PD biomarkers based on metabolomics techniques is of great significance for developing PD diagnosis. To comprehensively clarify the progress of current metabolic biomarker studies in PD, we reviewed 20 research articles regarding the discovery and validation of biomarkers for PD diagnosis from three mainstream academic databases (NIH PubMed, ISI Web of Science, and Elsevier ScienceDirect). By analyzing those materials, we summarized the metabolic biomarkers identified by those metabolomics studies and discussed the potential approaches used for biomarker verifications. In conclusion, this review provides a comprehensive and updated overview of PD metabolomics research in the past two decades and particularly discusses the validation of disease biomarkers. We hope those discussions might provide inspiration for PD biomarker discovery and verification in the future.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Xiaoying Fan
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Hongtian Yang
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China
| | - Yufeng Liu
- School of Pharmaceutical Sciences, Liaoning University, No. 66 Chongshan Middle Road, Huanggu District, Liaoning Province, 110036, Shenyang, People's Republic of China. .,Natural Products Pharmaceutical Engineering Technology Research Center of Liaoning Province, Shenyang, 110036, People's Republic of China.
| |
Collapse
|
14
|
Wang XT, Yu H, Liu FT, Zhang C, Ma YH, Wang J, Dong Q, Tan L, Wang H, Yu JT. Associations of sleep disorders with cerebrospinal fluid α-synuclein in prodromal and early Parkinson's disease. J Neurol 2021; 269:2469-2478. [PMID: 34605986 DOI: 10.1007/s00415-021-10812-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Our aim is to investigate the associations of sleep disorders with cerebrospinal fluid (CSF) α-synuclein (α-syn) in healthy controls (HCs), and patients with prodromal and early Parkinson's disease (PD). METHODS We included a total of 575 individuals, consisting of 360 PD individuals, 46 prodromal PD individuals, and 169 HCs. Multiple linear regression models and linear mixed-effects models were used to investigate the associations of sleep disorders with baseline and longitudinal CSF α-syn. Associations between the change rates of sleep disorders and CSF α-syn were further investigated via multiple linear regression models. RESULTS In PD, probable Rapid-eye-movement sleep Behavior Disorder (pRBD) (β = - 0.1199; P = 0.0444) and RBD sub-items, such as aggressive dreams (β = - 0.1652; P = 0.0072) and hurting bed partner (β = - 0.2468; P = 0.0010), contributed to lower CSF α-syn. The association between aggressive dreams and lower CSF α-syn further survived Bonferroni correction (P < 0.0036). In prodromal PD, dream-enacting (a specific RBD behavior) was significantly associated with decreased CSF α-syn during the follow-up (β = - 0.0124; P = 0.0237). HCs with daytime sleepiness when inactive-sitting in public places (β = - 0.0033; P = 0.0135) showed decreased CSF α-syn. Furthermore, increased possibilities of daytime sleepiness when sitting and reading contributed to a greater decrease of CSF α-syn in HCs (β = - 196.8779; P = 0.0433). CONCLUSIONS Sleep disorders were associated with decreased CSF α-syn. Sleep management may be important for disease monitoring and preventing the progression of α-syn pathology.
Collapse
Affiliation(s)
- Xiao-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Huan Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Feng-Tao Liu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Can Zhang
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Jian Wang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Han Wang
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
15
|
Henderson AR, Wang Q, Meechoovet B, Siniard AL, Naymik M, De Both M, Huentelman MJ, Caselli RJ, Driver-Dunckley E, Dunckley T. DNA Methylation and Expression Profiles of Whole Blood in Parkinson's Disease. Front Genet 2021; 12:640266. [PMID: 33981329 PMCID: PMC8107387 DOI: 10.3389/fgene.2021.640266] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common age-related neurodegenerative disease. It is presently only accurately diagnosed at an advanced stage by a series of motor deficits, which are predated by a litany of non-motor symptoms manifesting over years or decades. Aberrant epigenetic modifications exist across a range of diseases and are non-invasively detectable in blood as potential markers of disease. We performed comparative analyses of the methylome and transcriptome in blood from PD patients and matched controls. Our aim was to characterize DNA methylation and gene expression patterns in whole blood from PD patients as a foundational step toward the future goal of identifying molecular markers that could predict, accurately diagnose, or track the progression of PD. We found that differentially expressed genes (DEGs) were involved in the processes of transcription and mitochondrial function and that PD methylation profiles were readily distinguishable from healthy controls, even in whole-blood DNA samples. Differentially methylated regions (DMRs) were functionally varied, including near transcription factor nuclear transcription factor Y subunit alpha (NFYA), receptor tyrosine kinase DDR1, RING finger ubiquitin ligase (RNF5), acetyltransferase AGPAT1, and vault RNA VTRNA2-1. Expression quantitative trait methylation sites were found at long non-coding RNA PAX8-AS1 and transcription regulator ZFP57 among others. Functional epigenetic modules were highlighted by IL18R1, PTPRC, and ITGB2. We identified patterns of altered disease-specific DNA methylation and associated gene expression in whole blood. Our combined analyses extended what we learned from the DEG or DMR results alone. These studies provide a foundation to support the characterization of larger sample cohorts, with the goal of building a thorough, accurate, and non-invasive molecular PD biomarker.
Collapse
Affiliation(s)
- Adrienne R Henderson
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Qi Wang
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Bessie Meechoovet
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Ashley L Siniard
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Marcus Naymik
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Matthew De Both
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | | | | | - Travis Dunckley
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
16
|
Parkinson's disease and the non-motor symptoms: hyposmia, weight loss, osteosarcopenia. Aging Clin Exp Res 2020; 32:1211-1218. [PMID: 31989535 DOI: 10.1007/s40520-020-01470-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022]
Abstract
Non-motor symptoms (NMSs) are common in Parkinson's disease (PD) and can precede, sometimes for several years. NMSs include, other than gastrointestinal symptoms like constipation and dysphagia, also hyposmia, weight loss and osteosarcopenia. These three NMSs seem to be inter-related and affect patients' health and quality of life. Unfortunately, patients with these symptoms usually are not initially seen by a neurologist, and by the time they are consulted, nearly ~ 80% of the dopaminergic neurons in the substantia nigra have died. To date, no guidelines exist for screening, assessment and management of NMSs in general. A better understanding of these specific NMSs, likely in the context of others, will make it possible to approach and optimise the treatment of the motor symptoms thereby enhancing the welfare of PD patients. Identifying the NMSs could be very helpful, and among them, hyposmia, weight loss and osteosarcopenia may play an important role in solving the limitations in the diagnosis of PD. A strict collaboration between general practitioners, clinicians, geriatricians and neurologists can be one approach towards the diagnosis of pre-PD. Waiting until the motor symptoms develop and the patient is finally visited by the neurologist could be too late, considering the catastrophic prognosis of the disease.
Collapse
|
17
|
Ugrumov M. Development of early diagnosis of Parkinson's disease: Illusion or reality? CNS Neurosci Ther 2020; 26:997-1009. [PMID: 32597012 PMCID: PMC7539842 DOI: 10.1111/cns.13429] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
The fight against neurodegenerative diseases, Alzheimer disease and Parkinson's disease (PD), is a challenge of the 21st century. The low efficacy of treating patients is due to the late diagnosis and start of therapy, after the degeneration of most specific neurons and depletion of neuroplasticity. It is believed that the development of early diagnosis (ED) and preventive treatment will delay the onset of specific symptoms. This review evaluates methodologies for developing ED of PD. Since PD is a systemic disease, and the degeneration of certain neurons precedes that of nigrostriatal dopaminergic neurons that control motor function, the current methodology is based on searching biomarkers, such as premotor symptoms and changes in body fluids (BF) in patients. However, all attempts to develop ED were unsuccessful. Therefore, it is proposed to enhance the current methodology by (i) selecting among biomarkers found in BF in patients at the clinical stage those that are characteristics of animal models of the preclinical stage, (ii) searching biomarkers in BF in subjects at the prodromal stage, selected by detecting premotor symptoms and failure of the nigrostriatal dopaminergic system. Moreover, a new methodology was proposed for the development of ED of PD using a provocative test, which is successfully used in internal medicine.
Collapse
Affiliation(s)
- Michael Ugrumov
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology RAS, Moscow, Russia
| |
Collapse
|
18
|
Hornung S, Dutta S, Bitan G. CNS-Derived Blood Exosomes as a Promising Source of Biomarkers: Opportunities and Challenges. Front Mol Neurosci 2020; 13:38. [PMID: 32265650 PMCID: PMC7096580 DOI: 10.3389/fnmol.2020.00038] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Eukaryotic cells release different types of extracellular vesicles (EVs) including exosomes, ectosomes, and microvesicles. Exosomes are nanovesicles, 30–200 nm in diameter, that carry cell- and cell-state-specific cargo of proteins, lipids, and nucleic acids, including mRNA and miRNA. Recent studies have shown that central nervous system (CNS)-derived exosomes may carry amyloidogenic proteins and facilitate their cell-to-cell transfer, thus playing a critical role in the progression of neurodegenerative diseases, such as tauopathies and synucleinopathies. CNS-derived exosomes also have been shown to cross the blood-brain-barrier into the bloodstream and therefore have drawn substantial attention as a source of biomarkers for various neurodegenerative diseases as they can be isolated via a minimally invasive blood draw and report on the biochemical status of the CNS. However, although isolating specific brain-cell-derived exosomes from the blood is theoretically simple and the approach has great promise, practical details are of crucial importance and may compromise the reproducibility and utility of this approach, especially when different laboratories use different protocols. In this review we discuss the role of exosomes in neurodegenerative diseases, the usefulness of CNS-derived blood exosomes as a source of biomarkers for these diseases, and practical challenges associated with the methodology of CNS-derived blood exosomes and subsequent biomarker analysis.
Collapse
Affiliation(s)
- Simon Hornung
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Suman Dutta
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
19
|
Parkinson's disease prognostic scores for progression of cognitive decline. Sci Rep 2019; 9:17485. [PMID: 31767922 PMCID: PMC6877592 DOI: 10.1038/s41598-019-54029-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/08/2019] [Indexed: 11/08/2022] Open
Abstract
Clinical and biochemical diversity of Parkinson’s disease (PD) presents a major challenge for accurate diagnosis and prediction of its progression. We propose, develop and optimize PD clinical scores as efficient integrated progression biomarkers for prediction of the likely rate of cognitive decline in PD patients. We considered 269 drug-naïve participants from the Parkinson’s Progression Marker Initiative database, diagnosed with idiopathic PD and observed between 4 and 6 years. Nineteen baseline clinical and pathological measures were systematically considered. Relative variable importance and logistic regressions were used to optimize combinations of significant baseline measures as integrated biomarkers. Parkinson’s disease cognitive decline scores were designed as new clinical biomarkers using optimally categorized baseline measures. Specificities and sensitivities of the biomarkers reached ~93% for prediction of severe rate of cognitive decline (with more than 5 points decline in 4 years on the Montreal Cognitive Assessment scale), and up to ~73% for mild-to-moderate decline (between 1 and 5 points decline). The developed biomarkers and clinical scores could resolve the long-standing clinical problem about reliable prediction of PD progression into cognitive deterioration. The outcomes also provide insights into the contributions of individual clinical and pathological measures to PD progression, and will assist with better-targeted treatment regiments, stratification of clinical trial and their evaluation.
Collapse
|
20
|
Zeighami Y, Fereshtehnejad SM, Dadar M, Collins DL, Postuma RB, Dagher A. Assessment of a prognostic MRI biomarker in early de novo Parkinson's disease. Neuroimage Clin 2019; 24:101986. [PMID: 31514113 PMCID: PMC6742805 DOI: 10.1016/j.nicl.2019.101986] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 07/29/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Commonly used neuroimaging biomarkers in Parkinson's disease (PD) are useful for diagnosis but poor at predicting outcomes. We explored whether an atrophy pattern from whole-brain structural MRI, measured in the drug-naïve early stage, could predict PD prognosis. METHODS 362 de novo PD patients with T1-weighted MRI (n = 222 for the main analysis, 140 for the validation analysis) were recruited from the Parkinson's Progression Markers Initiative (PPMI). We investigated a previously identified PD-specific network atrophy pattern as a potential biomarker of disease severity and prognosis. Progression trajectories of motor function (MDS-UPDRS-part III), cognition (Montreal Cognitive Assessment (MoCA)), and a global composite outcome measure were compared between atrophy tertiles using mixed effect models. The prognostic value of the MRI atrophy measure was compared with 123I ioflupane single photon emission computed tomography, the postural-instability-gait-disturbance score, and cerebrospinal fluid markers. FINDINGS After 4.5 years follow-up, PD-specific atrophy network score at baseline significantly predicted change in UPDRS-part III (r = -0.197, p = .003), MoCA (r = 0.253, p = .0002) and global composite outcome (r = -0.249, p = .0002). Compared with the 3rd tertile (i.e. least atrophy), the tertile with the highest baseline atrophy (i.e. the 1st tertile) had a 3-point annual faster progression in UPDRS-part III (p = .012), faster worsening of posture-instability gait scores (+0.21 further annual increase, p < .0001), faster decline in MoCA (-0.74 further annual decline in MoCA, p = .0372) and a + 0.38 (p = .0029) faster annual increase in the global composite z-score. All findings were replicated in a validation analysis using 1.5T MRI. Receiver operating characteristic analysis confirmed the superiority of the MRI biomarker, although it had modest AUC values (0.63). By comparison, the other biomarkers were limited in their ability to predict prognosis either in the main or validation analysis. INTERPRETATION A PD-specific network atrophy pattern predicts progression of motor, cognitive, and global outcome in PD, and is a better predictor of prognosis than any of the other tested biomarkers. Therefore, it has potential as a prognostic biomarker for clinical trials of early PD.
Collapse
Affiliation(s)
- Yashar Zeighami
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Seyed-Mohammad Fereshtehnejad
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Mahsa Dadar
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - D Louis Collins
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ronald B Postuma
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Pizarro C, Esteban-Díez I, Espinosa M, Rodríguez-Royo F, González-Sáiz JM. An NMR-based lipidomic approach to identify Parkinson's disease-stage specific lipoprotein-lipid signatures in plasma. Analyst 2019; 144:1334-1344. [PMID: 30564825 DOI: 10.1039/c8an01778f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Disturbances in lipid composition and lipoproteins metabolism can play a crucial role in the pathogenesis of Parkinson's disease (PD) and other neurodegenerative diseases. The lipidomic strategy proposed here involves lipoprotein profiling using NMR spectroscopy and multivariate data pre-processing and analysis tools on 94 plasma samples (belonging to 38 early-stage PD patients, 10 PD-related dementia patients, 23 persons with Alzheimer's dementia, and 23 healthy control subjects) to firstly differentiate PD patients (irrespective of the stage of the disease) from persons with Alzheimer's disease (AD) as well as from controls, and then to discriminate among PD patients according to disease severity. The whole data set was subdivided into 86 training and 8 external test samples for validation purposes. A two-step classification scheme, based on linear discriminant analysis with variable selection accomplished by a stepwise orthogonalisation procedure, was proposed to optimise classification performance. Careful pre-processing of NMR signals was crucial to ensure data set quality. A total of 30 chemical shift buckets enabled differentiation between PD patients (regardless of disease severity), AD and control subjects, providing classification, cross-validation and external prediction rates of 100% in all cases. Only 15 variables were required to further discriminate between early-stage PD and PD-related dementia, again with 100% correct classifications, and internal/external predictions. The simplicity and effectiveness of the classification methodology proposed support the use of NMR spectroscopy, in combination with chemometrics, as a viable alternative diagnostic tool to conventional PD clinical diagnosis.
Collapse
Affiliation(s)
- Consuelo Pizarro
- Department of Chemistry, University of La Rioja, E-26006 Logroño, Spain.
| | | | | | | | | |
Collapse
|
22
|
D'Andrea G, Pizzolato G, Gucciardi A, Stocchero M, Giordano G, Baraldi E, Leon A. Different Circulating Trace Amine Profiles in De Novo and Treated Parkinson's Disease Patients. Sci Rep 2019; 9:6151. [PMID: 30992490 PMCID: PMC6467876 DOI: 10.1038/s41598-019-42535-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Early diagnosis of Parkinson’s disease (PD) remains a challenge to date. New evidence highlights the potential clinical value of circulating trace amines (TAs) in early-stage PD and their involvement in disease progression. A new ultra performance chromatography mass spectrometry (UPLC-MS/MS) method was developed to quantify plasmatic TAs, and the catecholamines and indolamines pertaining to the same biochemical pathways. Three groups of subjects were recruited: 21 de novo, drug untreated, PD patients, 27 in treatment PD patients and 10 healthy subjects as controls. Multivariate and univariate data analyses were applied to reveal metabolic changes among the groups in attempt to discover new putative markers for early PD detection and disease progression. Different circulating levels of tyrosine (p = 0.002), tyramine (p < 0.001), synephrine (p = 0.015), norepinephrine (p = 0.012), metanephrine (p = 0.001), β-phenylethylamine (p = 0.001) and serotonin (p = 0.006) were found among the three groups. While tyramine behaves as a putative biomarker for early-stage PD (AUC = 0.90) tyramine, norepinephrine, and tyrosine appear to act as biomarkers of disease progression (AUC > 0.75). The findings of this pilot cross-sectional study suggest that biochemical anomalies of the aminergic and indolic neurotransmitters occur in PD patients. Compounds within the TAs family may constitute putative markers for early stage detection and progression of PD.
Collapse
Affiliation(s)
| | - Gilberto Pizzolato
- Department of Medical Sciences, Neurology Unit, University of Trieste, Trieste, Italy
| | - Antonina Gucciardi
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy. .,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy.
| | - Matteo Stocchero
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Giuseppe Giordano
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Eugenio Baraldi
- Mass Spectrometry and Metabolomic Laboratory, Women's and Children's Health Department, University of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Cittàdella Speranza, Padova, Italy
| | - Alberta Leon
- Research and Innovation (R&I Genetics) s.r.l., Padova, Italy
| |
Collapse
|
23
|
Zhao ZH, Chen ZT, Zhou RL, Zhang X, Ye QY, Wang YZ. Increased DJ-1 and α-Synuclein in Plasma Neural-Derived Exosomes as Potential Markers for Parkinson's Disease. Front Aging Neurosci 2019; 10:438. [PMID: 30692923 PMCID: PMC6339871 DOI: 10.3389/fnagi.2018.00438] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/21/2018] [Indexed: 01/12/2023] Open
Abstract
The diagnosis of PD might be in difficulty, especially in the early stages. Therefore, the identification of novel biomarkers is imperative for the diagnosis and monitoring disease progression in PD. DJ-1 and α-synuclein, are two proteins that are critically involved in the pathogenesis of PD, and they have been examined as disease biomarkers in studies. However, no study exists regarding DJ-1 in plasma neural-derived exosomes. In the present study, the levels of DJ-1 and α-synuclein in plasma neural-derived exosomes were studied together in order to investigate novel biomarkers for PD. DJ-1 and α-synuclein in plasma and plasma neural-derived exosomes of the patients with PD and controls were quantified by ELISAs. The data revealed that the levels of DJ-1 and α-synuclein in plasma neural-derived exosomes and the ratio of plasma neural-derived exosomal DJ-1 to total DJ-1 were significantly higher in patients with PD, compared with controls, while levels of the two proteins in plasma exhibited no difference between the patients with PD and controls. However, no relationship was identified between biomarkers and disease progression. In addition, significant positive correlations between DJ-1 and α-synuclein in plasma neural-derived exosomes were found in the patients with PD and in healthy individuals. We hypothesize that DJ-1 in plasma neural-derived exosomes may be used as a potential biomarker as α-synuclein in PD and they might participate in the mechanism of PD together.
Collapse
Affiliation(s)
- Zhen-Hua Zhao
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Zhi-Ting Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui-Ling Zhou
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Xu Zhang
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Qin-Yong Ye
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yin-Zhou Wang
- Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
24
|
He R, Yan X, Guo J, Xu Q, Tang B, Sun Q. Recent Advances in Biomarkers for Parkinson's Disease. Front Aging Neurosci 2018; 10:305. [PMID: 30364199 PMCID: PMC6193101 DOI: 10.3389/fnagi.2018.00305] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 09/14/2018] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is one of the common progressive neurodegenerative disorders with several motor and non-motor symptoms. Most of the motor symptoms may appear at a late stage where most of the dopaminergic neurons have been already damaged. In order to provide better clinical intervention and treatment at the onset of disease, it is imperative to find accurate biomarkers for early diagnosis, including prodromal diagnosis and preclinical diagnosis. At the same time, these reliable biomarkers can also be utilized to monitor the progress of the disease. In this review article, we will discuss recent advances in the development of PD biomarkers from different aspects, including clinical, biochemical, neuroimaging and genetic aspects. Although various biomarkers for PD have been developed so far, their specificity and sensitivity are not ideal when applied individually. So, the combination of multimodal biomarkers will greatly improve the diagnostic accuracy and facilitate the implementation of personalized medicine.
Collapse
Affiliation(s)
- Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qiying Sun
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Cova I, Priori A. Diagnostic biomarkers for Parkinson's disease at a glance: where are we? J Neural Transm (Vienna) 2018; 125:1417-1432. [PMID: 30145631 PMCID: PMC6132920 DOI: 10.1007/s00702-018-1910-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder whose aetiology remains unclear: degeneration involves several neurotransmission systems, resulting in a heterogeneous disease characterized by motor and non-motor symptoms. PD causes progressive disability that responds only to symptomatic therapies. Future advances include neuroprotective strategies for use in at-risk populations before the clinical onset of disease, hence the continuing need to identify reliable biomarkers that can facilitate the clinical diagnosis of PD. In this evaluative review, we summarize information on potential diagnostic biomarkers for use in the clinical and preclinical stages of PD.
Collapse
Affiliation(s)
- Ilaria Cova
- Neurology Unit, L. Sacco University Hospital, Milan, Italy
| | - Alberto Priori
- Department of Health Sciences, "Aldo Ravelli" Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan and ASST Santi Paolo e Carlo, Milan, Italy.
| |
Collapse
|
26
|
Kim A, Nigmatullina R, Zalyalova Z, Soshnikova N, Krasnov A, Vorobyeva N, Georgieva S, Kudrin V, Narkevich V, Ugrumov M. Upgraded Methodology for the Development of Early Diagnosis of Parkinson's Disease Based on Searching Blood Markers in Patients and Experimental Models. Mol Neurobiol 2018; 56:3437-3450. [PMID: 30128652 DOI: 10.1007/s12035-018-1315-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 08/10/2018] [Indexed: 01/08/2023]
Abstract
Numerous attempts to develop an early diagnosis of Parkinson's disease (PD) by searching biomarkers in biological fluids were unsuccessful. The drawback of this methodology is searching markers in patients at the clinical stage without guarantee that they are also characteristic of either preclinical stage or prodromal stage (preclinical-prodromal stage). We attempted to upgrade this methodology by selecting only markers that are found both in patients and in PD animal models. HPLC and RT-PCR were used to estimate the concentration of amino acids, catecholamines/metabolites in plasma and gene expression in lymphocytes in 36 untreated early-stage PD patients and 52 controls, and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice at modeling the clinical ("symptomatic") stage and preclinical-prodromal ("presymptomatic") stage of PD. It was shown that among 13 blood markers found in patients, 7 markers are characteristic of parkinsonian symptomatic mice and 3 markers of both symptomatic and presymptomatic mice. According to our suggestion, the detection of the same marker in patients and symptomatic animals indicates adequate reproduction of pathogenesis along the corresponding metabolic pathway, whereas the detection of the same marker in presymptomatic animals indicates its specificity for preclinical-prodromal stage. This means that the minority of markers found in patients-decreased concentration of L-3,4-dihydroxyphenylalanine (L-DOPA) and dihydroxyphenylacetic acid (DOPAC) and increased dopamine D3 receptor gene expression-are specific for preclinical-prodromal stage and are suitable for early diagnosis of PD. Thus, we upgraded a current methodology for development of early diagnosis of PD by searching blood markers not only in patients but also in parkinsonian animals.
Collapse
Affiliation(s)
- Alexander Kim
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Razina Nigmatullina
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia
| | - Zuleikha Zalyalova
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia
- Kazan Hospital for War Veterans, Ministry of Health of the Republic of Tatarstan, Kazan, Russia
| | | | - Alexey Krasnov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Sofia Georgieva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | - Michael Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
- National Research University Higher School of Economics, Moscow, Russia.
| |
Collapse
|
27
|
Kohler I, Hankemeier T, van der Graaf PH, Knibbe CA, van Hasselt JC. Integrating clinical metabolomics-based biomarker discovery and clinical pharmacology to enable precision medicine. Eur J Pharm Sci 2017; 109S:S15-S21. [DOI: 10.1016/j.ejps.2017.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022]
|
28
|
Yu Z, Zhang S, Wang D, Fan M, Gao F, Sun W, Li Z, Li S. The significance of uric acid in the diagnosis and treatment of Parkinson disease: An updated systemic review. Medicine (Baltimore) 2017; 96:e8502. [PMID: 29137045 PMCID: PMC5690738 DOI: 10.1097/md.0000000000008502] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/04/2017] [Accepted: 10/08/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Parkinson disease (PD) is a neurodegenerative disease characterized by chronic and progressive loss of dopaminergic neurons in substansia nigra pars compacta. Oxidative stress is proposed to play a critical role in the pathogenesis of PD. Uric acid (UA), as an important physiological antioxidant, is identified a molecular predictor associated with a decreased risk and a slower disease progression for PD and potential neuroprotectant of PD by increasing epidemiological and clinical evidences. Within this review, we will present a comprehensive overview of the data linking UA to PD in recent years. METHODS We searched PubMed, EMBASE, Web of Science databases for relevant studies. Any observational or experimental studies that evaluated UA and PD were our goal of searching the electric databases. RESULTS Twelve studies that evaluated UA and PD were identified in this review. We reviewed the roles of UA in the pathogenesis of PD, the association of UA with morbidity, severity/progression, nonmotor symptoms, motor complications of PD, with an attempt to provide new ideas for diagnosis and treatment in PD. CONCLUSION Our findings supported that lots of clinical and epidemiological data observed lower UA levels in PD patients. Manipulation of UA or its precursors' concentration could be effective to treat or prevent PD. However, it is still suspectable that higher UA levels are better enough to PD patients. Furthermore, for the complex nature of PD and its heterogeneous genetic and environmental influences, it is inadequate for just manipulating UA in treating the disease.
Collapse
Affiliation(s)
- Zhange Yu
- Department of Acupuncture, China-Japan Friendship Hospital, Beijing
| | - Shuai Zhang
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province
| | - Dongdong Wang
- Department of Orthopedics, Tumd Right Banner Hospital, Baotou City
| | - Meng Fan
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Fuqiang Gao
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Wei Sun
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Zirong Li
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Shiliang Li
- Department of Acupuncture, China-Japan Friendship Hospital, Beijing
| |
Collapse
|
29
|
Monnot C, Zhang X, Nikkhou-Aski S, Damberg P, Svenningsson P. Asymmetric dopaminergic degeneration and levodopa alter functional corticostriatal connectivity bilaterally in experimental parkinsonism. Exp Neurol 2017; 292:11-20. [PMID: 28223037 PMCID: PMC5405850 DOI: 10.1016/j.expneurol.2017.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/02/2017] [Accepted: 02/17/2017] [Indexed: 12/11/2022]
Abstract
Asymmetric dopamine loss is commonly found in early Parkinson's disease (PD), but its effects on functional networks have been difficult to delineate in PD patients because of variations in age, disease duration and therapy. Here we used unilateral 6-hydroxydopamine-lesioned (6-OHDA) rats and controls and treated them with a single intraperitoneal injection of levodopa (L-DOPA) before performing diffusion weighted MRI and resting state functional MRI (rs-fMRI). In accordance with a neurodegeneration of the nigrostriatal dopaminergic pathway, diffusion tensor imaging showed increased radial diffusivity and decreased fractional anisotropy in the lesioned substantia nigra. Likewise a deterministic connectometry approach showed increase of isotropic diffusion values in the medial forebrain bundle. rs-fMRI showed reduced interhemispheric functional connectivity (FC) between the intact and the 6-OHDA lesioned caudate-putamen. Unexpectedly, there was an increased FC between the 6-OHDA lesioned caudate-putamen and sensorimotor cortices of both hemispheres. L-DOPA reversed the FC changes between the dopamine denervated caudate-putamen and the sensorimotor cortices, but not the reduced interhemispheric FC between caudate-putamina. Similarly, L-DOPA induced c-fos expression in both sensorimotor cortices, but only in the dopamine-depleted caudate-putamen. Taken together, these data suggest that asymmetric degeneration of the nigrostriatal dopamine pathway results in functional asynchrony between the intact and 6-OHDA-lesioned caudate-putamen and increased interhemispheric synchrony between sensorimotor cortices. The results also indicate that the initial effect of L-DOPA is to restore functional corticostriatal connectivity rather than synchronize caudate-putamina. Rats unilaterally lesioned with 6-hydroxydopamine (6-OHDA) are examined using MRI. Diffusion MRI revealed loss of fractional anisotropy in a lesioned substantia nigra. rs-fMRI showed lower functional connectivity (FC) btw intact and lesioned striata. FC increased between the lesioned striatum and both sensorimotor cortices. Levodopa normalized FC between sensorimotor cortices and lesioned striatum.
Collapse
Affiliation(s)
- Cyril Monnot
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Sahar Nikkhou-Aski
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Peter Damberg
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
30
|
Abstract
INTRODUCTION Parkinson's disease (PD) is an insidious disorder affecting more than 1-2% of the population over the age of 65. Understanding the etiology of PD may create opportunities for developing new treatments. Genomic and transcriptomic studies are useful, but do not provide evidence for the actual status of the disease. Conversely, proteomic studies deal with proteins, which are real time players, and can hence provide information on the dynamic nature of the affected cells. The number of publications relating to the proteomics of PD is vast. Therefore, there is a need to evaluate the current proteomics literature and establish the connections between the past and the present to foresee the future. Areas covered: PubMed and Web of Science were used to retrieve the literature associated with PD proteomics. Studies using human samples, model organisms and cell lines were selected and reviewed to highlight their contributions to PD. Expert commentary: The proteomic studies associated with PD achieved only limited success in facilitating disease diagnosis, monitoring and progression. A global system biology approach using new models is needed. Future research should integrate the findings of proteomics with other omics data to facilitate both early diagnosis and the treatment of PD.
Collapse
Affiliation(s)
- Murat Kasap
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| | - Gurler Akpinar
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| | - Aylin Kanli
- a Department of Medical Biology/DEKART Proteomics Laboratory , Kocaeli University Medical School , Kocaeli , Turkey
| |
Collapse
|
31
|
Alterations of p11 in brain tissue and peripheral blood leukocytes in Parkinson's disease. Proc Natl Acad Sci U S A 2017; 114:2735-2740. [PMID: 28137881 DOI: 10.1073/pnas.1621218114] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Individuals with Parkinson's disease (PD) often suffer from comorbid depression. P11 (S100A10), a member of the S100 family of proteins, is expressed widely throughout the body and is involved in major depressive disorder and antidepressant response. Central p11 levels are reduced in postmortem tissue from depressed individuals; however, p11 has not yet been investigated in PD patients with depression or those without depression. We investigated p11 levels in postmortem PD brains and assessed whether peripheral p11 levels correlate with disease severity. Substantia nigra, putamen, and cortical p11 protein levels were assessed in postmortem brain samples from PD patients and matched controls. In a different set of postmortem brains, p11 mRNA expression was measured in dopaminergic cells from the substantia nigra. Both p11 protein and mRNA levels were decreased in PD patients. Peripheral p11 protein levels were investigated in distinct leukocyte populations from PD patients with depression and those without depression. Monocyte, natural killer (NK) cell, and cytotoxic T-cell p11 levels were positively associated with the severity of PD, and NK cell p11 levels were positively associated with depression scores. Given that inflammation plays a role in both PD and depression, it is intriguing that peripheral p11 levels are altered in immune cells in both conditions. Our data provide insight into the pathological alterations occurring centrally and peripherally in PD. Moreover, if replicated in other cohorts, p11 could be an easily accessible biomarker for monitoring the severity of PD, especially in the context of comorbid depression.
Collapse
|
32
|
Guimarães RP, Arci Santos MC, Dagher A, Campos LS, Azevedo P, Piovesana LG, De Campos BM, Larcher K, Zeighami Y, Scarparo Amato-Filho AC, Cendes F, D'Abreu ACF. Pattern of Reduced Functional Connectivity and Structural Abnormalities in Parkinson's Disease: An Exploratory Study. Front Neurol 2017; 7:243. [PMID: 28133455 PMCID: PMC5233672 DOI: 10.3389/fneur.2016.00243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/21/2016] [Indexed: 02/03/2023] Open
Abstract
Background MRI brain changes in Parkinson’s disease (PD) are controversial. Objectives We aimed to describe structural and functional changes in PD. Methods Sixty-six patients with PD (57.94 ± 10.25 years) diagnosed according to the UK Brain Bank criteria were included. We performed a whole brain analysis using voxel-based morphometry (VBM–SPM 8 software), cortical thickness (CT) using CIVET, and resting-state fMRI using the Neuroimaging Analysis Kit software to compare patients and controls. For VBM and CT we classified subjects into three groups according to disease severity: mild PD [Hoehn and Yahr scale (HY) 1–1.5], moderate PD (HY 2–2.5), and severe PD (HY 3–5). Results We observed gray matter atrophy in the insula and inferior frontal gyrus in the moderate PD and in the insula, frontal gyrus, putamen, cingulated, and paracingulate gyri in the severe groups. In the CT analysis, in mild PD, cortical thinning was restricted to the superior temporal gyrus, gyrus rectus, and olfactory cortex; in the moderate group, the postcentral gyrus, supplementary motor area, and inferior frontal gyrus were also affected; in the severe PD, areas such as the precentral and postentral gyrus, temporal pole, fusiform, and occipital gyrus had reduced cortical thinning. We observed altered connectivity at the default mode, visual, sensorimotor, and cerebellar networks. Conclusion Subjects with mild symptoms already have cortical involvement; however, further cerebral involvement seems to follow Braak’s proposed mechanism. Similar regions are affected both structurally and functionally. We believe the combination of different MRI techniques may be useful in evaluating progressive brain involvement and they may eventually be used as surrogate markers of disease progression.
Collapse
Affiliation(s)
- Rachel Paes Guimarães
- Department of Neurology, University of Campinas, Campinas, Brazil; Laboratory of Neuroimaging, University of Campinas, Campinas, Brazil; Montreal Neurological Institute, Brain Imaging Center, McGill University, Montreal, QC, Canada
| | | | - Alain Dagher
- Montreal Neurological Institute, Brain Imaging Center, McGill University , Montreal, QC , Canada
| | | | - Paula Azevedo
- Department of Neurology, University of Campinas , Campinas , Brazil
| | | | | | - Kevin Larcher
- Montreal Neurological Institute, Brain Imaging Center, McGill University , Montreal, QC , Canada
| | - Yashar Zeighami
- Montreal Neurological Institute, Brain Imaging Center, McGill University , Montreal, QC , Canada
| | | | - Fernando Cendes
- Department of Neurology, University of Campinas, Campinas, Brazil; Laboratory of Neuroimaging, University of Campinas, Campinas, Brazil
| | - Anelyssa Cysne Frota D'Abreu
- Department of Neurology, University of Campinas, Campinas, Brazil; Laboratory of Neuroimaging, University of Campinas, Campinas, Brazil
| |
Collapse
|
33
|
Chaudhuri KR, Bhidayasiri R, van Laar T. Unmet needs in Parkinson's disease: New horizons in a changing landscape. Parkinsonism Relat Disord 2016; 33 Suppl 1:S2-S8. [PMID: 27932224 DOI: 10.1016/j.parkreldis.2016.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/22/2016] [Accepted: 11/25/2016] [Indexed: 11/15/2022]
Abstract
The success of levodopa and other classes of drugs have meant that most people with Parkinson's disease enjoy a good quality of life for many years. However, despite the availability of several drugs and formulations that can be used as monotherapy and in combination, there are a number of disease features that the current therapies are unable to address. The disease continues to progress despite treatment, patients suffer from a myriad of motor and non-motor symptoms, and a neuroprotective therapy is urgently required. To move forward with medical and surgical management, it is important to consider new insights that recent research offers and in this review we examine how a better understanding of the disease pathology and progression might improve and enrich our daily clinical practice. It is also timely to consider the service provision changes that will increasingly be needed to effectively manage the needs of the aging population.
Collapse
Affiliation(s)
- K Ray Chaudhuri
- The Maurice Wohl Clinical Neuroscience Institute, King's College London and National Parkinson Foundation Centre of Excellence, King's College Hospital London, UK
| | - Roongroj Bhidayasiri
- Chulalongkorn Center of Excellence for Parkinson's Disease & Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University, and King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Rehabilitation Medicine, Juntendo University, Tokyo, Japan.
| | - Teus van Laar
- Department of Neurology, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Marsh AG, Cottrell MT, Goldman MF. Epigenetic DNA Methylation Profiling with MSRE: A Quantitative NGS Approach Using a Parkinson's Disease Test Case. Front Genet 2016; 7:191. [PMID: 27853465 PMCID: PMC5090125 DOI: 10.3389/fgene.2016.00191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/14/2016] [Indexed: 11/22/2022] Open
Abstract
Epigenetics is a rapidly developing field focused on deciphering chemical fingerprints that accumulate on human genomes over time. As the nascent idea of precision medicine expands to encompass epigenetic signatures of diagnostic and prognostic relevance, there is a need for methodologies that provide high-throughput DNA methylation profiling measurements. Here we report a novel quantification methodology for computationally reconstructing site-specific CpG methylation status from next generation sequencing (NGS) data using methyl-sensitive restriction endonucleases (MSRE). An integrated pipeline efficiently incorporates raw NGS metrics into a statistical discrimination platform to identify functional linkages between shifts in epigenetic DNA methylation and disease phenotypes in samples being analyzed. In this pilot proof-of-concept study we quantify and compare DNA methylation in blood serum of individuals with Parkinson's Disease relative to matched healthy blood profiles. Even with a small study of only six samples, a high degree of statistical discrimination was achieved based on CpG methylation profiles between groups, with 1008 statistically different CpG sites (p < 0.0025, after false discovery rate correction). A methylation load calculation was used to assess higher order impacts of methylation shifts on genes and pathways and most notably identified FGF3, FGF8, HTT, KMTA5, MIR8073, and YWHAG as differentially methylated genes with high relevance to Parkinson's Disease and neurodegeneration (based on PubMed literature citations). Of these, KMTA5 is a histone methyl-transferase gene and HTT is Huntington Disease Protein or Huntingtin, for which there are well established neurodegenerative impacts. The future need for precision diagnostics now requires more tools for exploring epigenetic processes that may be linked to cellular dysfunction and subsequent disease progression.
Collapse
Affiliation(s)
- Adam G Marsh
- Center for Bioinformatics and Computational Biology, Delaware Biotechnology Institute, University of DelawareNewark, DE, USA; Genome Profiling LLC, Helen F. Graham Cancer Center and Research Institute, Center for Translational Cancer ResearchNewark, DE USA; Marine Biosciences, School of Marine Science and Policy, University of DelawareLewes, DE, USA
| | - Matthew T Cottrell
- Genome Profiling LLC, Helen F. Graham Cancer Center and Research Institute, Center for Translational Cancer ResearchNewark, DE USA; Marine Biosciences, School of Marine Science and Policy, University of DelawareLewes, DE, USA
| | - Morton F Goldman
- Genome Profiling LLC, Helen F. Graham Cancer Center and Research Institute, Center for Translational Cancer Research Newark, DE USA
| |
Collapse
|
35
|
Glutathione as a Biomarker in Parkinson's Disease: Associations with Aging and Disease Severity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9409363. [PMID: 27446510 PMCID: PMC4944065 DOI: 10.1155/2016/9409363] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/08/2016] [Indexed: 11/28/2022]
Abstract
Objectives. Oxidative stress contributes to Parkinson's disease (PD) pathophysiology and progression. The objective was to describe central and peripheral metabolites of redox metabolism and to describe correlations between glutathione (Glu) status, age, and disease severity. Methods. 58 otherwise healthy individuals with PD were examined during a single study visit. Descriptive statistics and scatterplots were used to evaluate normality and distribution of this cross-sectional sample. Blood tests and magnetic resonance spectroscopy (MRS) were used to collect biologic data. Spearman's rank-order correlation coefficients were used to evaluate the strength and direction of the association. The Unified PD Rating Scale (UPDRS) and the Patient-Reported Outcomes in PD (PRO-PD) were used to rate disease severity using regression analysis. Results. Blood measures of Glu decreased with age, although there was no age-related decline in MRS Glu. The lower the blood Glu concentration, the more severe the UPDRS (P = 0.02, 95% CI: −13.96, −1.14) and the PRO-PD (P = 0.01, 95% CI: −0.83, −0.11) scores. Discussion. These data suggest whole blood Glu may have utility as a biomarker in PD. Future studies should evaluate whether it is a modifiable risk factor for PD progression and whether Glu fortification improves PD outcomes.
Collapse
|