1
|
Wang X, Li W, Zhao X, Hu N, Wang X, Xiao X, Yang K, Sun T. Dysregulated Coagulation in Parkinson's Disease. Cells 2024; 13:1874. [PMID: 39594622 PMCID: PMC11592531 DOI: 10.3390/cells13221874] [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: 10/19/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD), a prevalent neurodegenerative disorder characterized by dopaminergic neuron degeneration and α-synuclein accumulation, has been increasingly associated with coagulation dysfunction. This review synthesizes emerging evidence linking dysregulated coagulation to PD pathophysiology. We examine the alterations in coagulation parameters, including elevated fibrinogen levels, impaired fibrinolysis, and platelet dysfunction, which collectively contribute to a hypercoagulable state in PD patients. Epidemiological studies have revealed a higher incidence of thrombotic events, such as deep vein thrombosis (DVT) and stroke, among PD patients, suggesting significant comorbidity between PD and coagulation disorders. This review explores the potential pathophysiological mechanisms underlying this association, focusing on the roles of inflammation and oxidative stress. Additionally, we discuss the limitations of current research and propose future directions. This comprehensive analysis underscores the importance of understanding the coagulation-neurodegeneration axis in PD, which may lead to novel diagnostic and therapeutic strategies for this debilitating condition.
Collapse
Affiliation(s)
- Xinqing Wang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China;
| | - Wenxin Li
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
| | - Ning Hu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
| | - Xi Wang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
| | - Xilin Xiao
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China;
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, China
| | - Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (W.L.); (X.Z.); (N.H.); (X.W.); (T.S.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
2
|
Sultana P, Novotny J. Clusterin: a double-edged sword in cancer and neurological disorders. EXCLI JOURNAL 2024; 23:912-936. [PMID: 39253532 PMCID: PMC11382300 DOI: 10.17179/excli2024-7369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 09/11/2024]
Abstract
Clusterin is a ubiquitously expressed glycoprotein that is involved in a whole range of biological processes. This protein is known to promote tumor survival and resistance to therapy in cancer, which contrasts sharply with its neuroprotective functions in various neurological diseases. This duality has led to recent investigations into the potential therapeutic applications of clusterin inhibition, particularly in cancer treatment. Inhibition of clusterin has been shown to be able to induce cancer cell senescence, suppress their growth and increase their sensitivity to therapy. The involvement of clusterin in the aging process makes its biological effects even more complex and offers a broad perspective for research and therapeutic exploration of various pathological conditions. This review critically examines the multiple functions of clusterin in cancer and neurological disorders and addresses the controversies surrounding its role in these areas. The assessment includes an in-depth analysis of the existing literature and examining the relationship of clusterin to fundamental aspects of cancer progression, including cell proliferation, apoptosis, metastasis, and drug resistance. In addition, the review addresses the neurobiological implications of clusterin and examines its controversial role in neuroprotection, neurodegeneration, and synaptic plasticity. Attention is also paid to the epigenetic regulation of clusterin expression. By clarifying conflicting findings and discrepancies in the literature, this review aims to provide a nuanced understanding of the molecular mechanisms underlying clusterin functions and its potential clinical implications in both cancer and neurodisorders. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Pinky Sultana
- Department of Physiology, Faculty of Science, Charles University, Prague 128 00, Czechia
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague 142 20, Czechia
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague 128 00, Czechia
| |
Collapse
|
3
|
Faizan M, Sachan N, Verma O, Sarkar A, Rawat N, Pratap Singh M. Cerebrospinal fluid protein biomarkers in Parkinson's disease. Clin Chim Acta 2024; 556:117848. [PMID: 38417781 DOI: 10.1016/j.cca.2024.117848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Proteomic profiling is an effective way to identify biomarkers for Parkinson's disease (PD). Cerebrospinal fluid (CSF) has direct connectivity with the brain and could be a source of finding biomarkers and their clinical implications. Comparative proteomic profiling has shown that a group of differentially displayed proteins exist. The studies performed using conventional and classical tools also supported the occurrence of these proteins. Many studies have highlighted the potential of CSF proteomic profiling for biomarker identification and their clinical applications. Some of these proteins are useful for disease diagnosis and prediction. Proteomic profiling of CSF also has immense potential to distinguish PD from similar neurodegenerative disorders. A few protein biomarkers help in fundamental knowledge generation and clinical interpretation. However, the specific biomarker of PD is not yet known. The use of proteomic approaches in clinical settings is also rare. A large-scale, multi-centric, multi-population and multi-continental study using multiple proteomic tools is warranted. Such a study can provide valuable, comprehensive and reliable information for a better understanding of PD and the development of specific biomarkers. The current article sheds light on the role of CSF proteomic profiling in identifying biomarkers of PD and their clinical implications. The article also explains the achievements, obstacles and hopes for future directions of this approach.
Collapse
Affiliation(s)
- Mohd Faizan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Nidhi Sachan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Oyashvi Verma
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Alika Sarkar
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Neeraj Rawat
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Capacity Building and Knowledge Services, ASSIST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.
| |
Collapse
|
4
|
Tiwari V, Shukla S. Lipidomics and proteomics: An integrative approach for early diagnosis of dementia and Alzheimer's disease. Front Genet 2023; 14:1057068. [PMID: 36845373 PMCID: PMC9946989 DOI: 10.3389/fgene.2023.1057068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and considered to be responsible for majority of worldwide prevalent dementia cases. The number of patients suffering from dementia are estimated to increase up to 115.4 million cases worldwide in 2050. Hence, AD is contemplated to be one of the major healthcare challenge in current era. This disorder is characterized by impairment in various signaling molecules at cellular and nuclear level including aggregation of Aβ protein, tau hyper phosphorylation altered lipid metabolism, metabolites dysregulation, protein intensity alteration etc. Being heterogeneous and multifactorial in nature, the disease do not has any cure or any confirmed diagnosis before the onset of clinical manifestations. Hence, there is a requisite for early diagnosis of AD in order to downturn the progression/risk of the disorder and utilization of newer technologies developed in this field are aimed to provide an extraordinary assistance towards the same. The lipidomics and proteomics constitute large scale study of cellular lipids and proteomes in biological matrices at normal stage or any stage of a disease. The study involves high throughput quantification and detection techniques such as mass spectrometry, liquid chromatography, nuclear mass resonance spectroscopy, fluorescence spectroscopy etc. The early detection of altered levels of lipids and proteins in blood or any other biological matrices could aid in preventing the progression of AD and dementia. Therefore, the present review is designed to focus on the recent techniques and early diagnostic criteria for AD, revealing the role of lipids and proteins in this disease and their assessment through different techniques.
Collapse
Affiliation(s)
- Virendra Tiwari
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing Biology, CSIR- Central Drug Research Institute, Lucknow, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,*Correspondence: Shubha Shukla,
| |
Collapse
|
5
|
The Emerging Roles of Extracellular Chaperones in Complement Regulation. Cells 2022; 11:cells11233907. [PMID: 36497163 PMCID: PMC9738919 DOI: 10.3390/cells11233907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
The immune system is essential to protect organisms from internal and external threats. The rapidly acting, non-specific innate immune system includes complement, which initiates an inflammatory cascade and can form pores in the membranes of target cells to induce cell lysis. Regulation of protein homeostasis (proteostasis) is essential for normal cellular and organismal function, and has been implicated in processes controlling immunity and infection. Chaperones are key players in maintaining proteostasis in both the intra- and extracellular environments. Whilst intracellular proteostasis is well-characterised, the role of constitutively secreted extracellular chaperones (ECs) is less well understood. ECs may interact with invading pathogens, and elements of the subsequent immune response, including the complement pathway. Both ECs and complement can influence the progression of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis, as well as other diseases including kidney diseases and diabetes. This review will examine known and recently discovered ECs, and their roles in immunity, with a specific focus on the complement pathway.
Collapse
|
6
|
Cervilla-Martínez JF, Rodríguez-Gotor JJ, Wypijewski KJ, Fontán-Lozano Á, Wang T, Santamaría E, Fuller W, Mejías R. Altered Cortical Palmitoylation Induces Widespread Molecular Disturbances in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms232214018. [PMID: 36430497 PMCID: PMC9696982 DOI: 10.3390/ijms232214018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
The relationship between Parkinson's disease (PD), the second-most common neurodegenerative disease after Alzheimer's disease, and palmitoylation, a post-translational lipid modification, is not well understood. In this study, to better understand the role of protein palmitoylation in PD and the pathways altered in this disease, we analyzed the differential palmitoyl proteome (palmitome) in the cerebral cortex of PD patients compared to controls (n = 4 per group). Data-mining of the cortical palmitome from PD patients and controls allowed us to: (i) detect a set of 150 proteins with altered palmitoylation in PD subjects in comparison with controls; (ii) describe the biological pathways and targets predicted to be altered by these palmitoylation changes; and (iii) depict the overlap between the differential palmitome identified in our study with protein interactomes of the PD-linked proteins α-synuclein, LRRK2, DJ-1, PINK1, GBA and UCHL1. In summary, we partially characterized the altered palmitome in the cortex of PD patients, which is predicted to impact cytoskeleton, mitochondrial and fibrinogen functions, as well as cell survival. Our study suggests that protein palmitoylation could have a role in the pathophysiology of PD, and that comprehensive palmitoyl-proteomics offers a powerful approach for elucidating novel cellular pathways modulated in this neurodegenerative disease.
Collapse
Affiliation(s)
- Juan F. Cervilla-Martínez
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Juan J. Rodríguez-Gotor
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
| | - Krzysztof J. Wypijewski
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- School of Life Sciences, University of Dundee, Dundee DD2 5DA, UK
| | - Ángela Fontán-Lozano
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Biomedicina de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n, 41013 Sevilla, Spain
| | - Tao Wang
- McKusick—Nathans Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Irunlarrea Street, 3, 31008 Pamplona, Spain
| | - William Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Rebeca Mejías
- Department of Physiology, School of Biology, University of Seville, Avenida de la Reina Mercedes, 6, 41012 Sevilla, Spain
- Instituto de Biomedicina de Sevilla, Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n, 41013 Sevilla, Spain
- Correspondence: ; Tel.: +34-954-559-549
| |
Collapse
|
7
|
Darweesh SK, De Vries NM, Helmich RC, Verbeek MM, Schwarzschild MA, Bloem BR. Inhibition of Neuroinflammation May Mediate the Disease-Modifying Effects of Exercise: Implications for Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1419-1422. [PMID: 35466957 PMCID: PMC9398068 DOI: 10.3233/jpd-223216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Sirwan K.L. Darweesh
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nienke M. De Vries
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rick C. Helmich
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M. Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Bastiaan R. Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Berdowska I, Matusiewicz M, Krzystek-Korpacka M. HDL Accessory Proteins in Parkinson’s Disease—Focusing on Clusterin (Apolipoprotein J) in Regard to Its Involvement in Pathology and Diagnostics—A Review. Antioxidants (Basel) 2022; 11:antiox11030524. [PMID: 35326174 PMCID: PMC8944556 DOI: 10.3390/antiox11030524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD)—a neurodegenerative disorder (NDD) characterized by progressive destruction of dopaminergic neurons within the substantia nigra of the brain—is associated with the formation of Lewy bodies containing mainly α-synuclein. HDL-related proteins such as paraoxonase 1 and apolipoproteins A1, E, D, and J are implicated in NDDs, including PD. Apolipoprotein J (ApoJ, clusterin) is a ubiquitous, multifunctional protein; besides its engagement in lipid transport, it modulates a variety of other processes such as immune system functionality and cellular death signaling. Furthermore, being an extracellular chaperone, ApoJ interacts with proteins associated with NDD pathogenesis (amyloid β, tau, and α-synuclein), thus modulating their properties. In this review, the association of clusterin with PD is delineated, with respect to its putative involvement in the pathological mechanism and its application in PD prognosis/diagnosis.
Collapse
Affiliation(s)
- Izabela Berdowska
- Correspondence: (I.B.); (M.M.); Tel.: +48-71-784-13-92 (I.B.); +48-71-784-13-70 (M.M.)
| | | | | |
Collapse
|
9
|
Ayton S, Hall S, Janelidze S, Kalinowski P, Palmqvist S, Belaidi AA, Roberts B, Roberts A, Stomrud E, Bush AI, Hansson O. The Neuroinflammatory Acute Phase Response in Parkinsonian-Related Disorders. Mov Disord 2022; 37:993-1003. [PMID: 35137973 DOI: 10.1002/mds.28958] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuroinflammation is implicated in the pathophysiology of Parkinson's disease (PD) and related conditions, yet prior clinical biomarker data report mixed findings. OBJECTIVES The aim was to measure a panel of neuroinflammatory acute phase response (APR) proteins in the cerebrospinal fluid (CSF) of participants with PD and related disorders. METHODS Eleven APR proteins were measured in the CSF of 867 participants from the BioFINDER cohort who were healthy (612) or had a diagnosis of PD (155), multiple system atrophy (MSA) (26), progressive supranuclear palsy (PSP) (22), dementia with Lewy bodies (DLB) (23), or Parkinson's disease with dementia (PDD) (29). RESULTS CSF APR proteins were mostly unchanged in PD, with only haptoglobin and α1-antitrypsin significantly elevated compared to controls. These proteins were variably increased in the other disorders. Certain protein components yielded unique signatures according to diagnosis: ferritin and transthyretin were selectively elevated in MSA and discriminated these patients from all others. Haptoglobin was selectively increased in PSP, discriminating this disease from MSA when used in combination with ferritin and transthyretin. This panel of proteins did not correlate well with severity of motor impairment in any disease category, but several (particularly ceruloplasmin and ferritin) were associated with memory performance (Mini-Mental State Examination) in patients with DLB and PDD. CONCLUSIONS These findings provide new insights into inflammatory changes in PD and related disorders while also introducing biomarkers of potential clinical diagnostic utility. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Abdel A Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Blaine Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anne Roberts
- Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
10
|
Ferroptosis as a Major Factor and Therapeutic Target for Neuroinflammation in Parkinson's Disease. Biomedicines 2021; 9:biomedicines9111679. [PMID: 34829907 PMCID: PMC8615560 DOI: 10.3390/biomedicines9111679] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence suggests that ferroptosis is not just a consequence but also a fundamental contributor to the development and progression of Parkinson’s disease (PD). Ferroptosis is characterized as iron-dependent regulated cell death caused by excessive lipid peroxidation, leading to plasma membrane rupture, release of damage-associated molecular patterns, and neuroinflammation. Due to the crucial role of intracellular iron in mediating the production of reactive oxygen species and the formation of lipid peroxides, ferroptosis is intimately controlled by regulators involved in many aspects of iron metabolism, including iron uptake, storage and export, and by pathways constituting the antioxidant systems. Translational and transcriptional regulation of iron homeostasis and redox status provide an integrated network to determine the sensitivity of ferroptosis. We herein review recent advances related to ferroptosis, ranging from fundamental mechanistic discoveries and cutting-edge preclinical animal studies, to clinical trials in PD and the regulation of neuroinflammation via ferroptosis pathways. Elucidating the roles of ferroptosis in the survival of dopaminergic neurons and microglial activity can enhance our understanding of the pathogenesis of PD and provide opportunities for the development of novel prevention and treatment strategies.
Collapse
|
11
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
12
|
Kaiserova M, Grambalova Z, Kurcova S, Otruba P, Prikrylova Vranova H, Mensikova K, Kanovsky P. Premotor Parkinson's disease: Overview of clinical symptoms and current diagnostic methods. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2021; 165:103-112. [PMID: 33542542 DOI: 10.5507/bp.2021.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/07/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is characterized by typical motor symptoms. However, recent studies show several non-motor features that may precede the development of the motor symptoms of PD. The best known premotor symptoms include hyposmia, REM sleep behavior disorder (RBD), constipation, and depression; other symptoms are excessive daytime somnolence, orthostatic hypotension and symptomatic hypotension, erectile or urinary dysfunction, musculoskeletal symptoms, pain, and global cognitive deficit. In this review, we summarize currently available diagnostic methods for these symptoms. We also briefly summarize neuroimaging, polyneuropathy, peripheral markers, and cerebrospinal fluid biomarkers that may be used in the early diagnosis of PD.
Collapse
Affiliation(s)
- Michaela Kaiserova
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Zuzana Grambalova
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Sandra Kurcova
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Pavel Otruba
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | | | - Katerina Mensikova
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Petr Kanovsky
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| |
Collapse
|
13
|
The Down-Regulation of Clusterin Expression Enhances the αSynuclein Aggregation Process. Int J Mol Sci 2020; 21:ijms21197181. [PMID: 33003328 PMCID: PMC7582711 DOI: 10.3390/ijms21197181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s Disease (PD) is a progressive neurodegenerative disease characterized by the presence of proteinaceous aggregates of αSynuclein (αSyn) in the dopaminergic neurons. Chaperones are key components of the proteostasis network that are able to counteract αSyn’s aggregation, as well as its toxic effects. Clusterin (CLU), a molecular chaperone, was consistently found to interfere with Aβ aggregation in Alzheimer’s Disease (AD). However, its role in PD pathogenesis has yet to be extensively investigated. In this study, we assessed the involvement of CLU in the αSyn aggregation process by using SH-SY5Y cells stably overexpressing αSyn (SH-Syn). First, we showed that αSyn overexpression caused a strong increase in CLU expression without affecting levels of Hsp27, Hsp70, and Hsp90, which are the chaperones widely recognized to counteract αSyn burden. Then, we demonstrated that αSyn aggregation, induced by proteasome inhibition, determines a strong increase of CLU in insoluble aggregates. Remarkably, we revealed that CLU down-regulation results in an increase of αSyn aggregates in SH-Syn without significantly affecting cell viability and the Unfolded Protein Response (UPR). Furthermore, we demonstrated the direct molecular interaction between CLU and αSyn via a co-immunoprecipitation (co-IP) assay. All together, these findings provide incontrovertible evidence that CLU is an important player in the response orchestrated by the cell to cope with αSyn burden.
Collapse
|
14
|
Katayama T, Sawada J, Takahashi K, Yahara O. Cerebrospinal Fluid Biomarkers in Parkinson's Disease: A Critical Overview of the Literature and Meta-Analyses. Brain Sci 2020; 10:brainsci10070466. [PMID: 32698474 PMCID: PMC7407121 DOI: 10.3390/brainsci10070466] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disorder; however, well-established biochemical markers have not yet been identified. This review article covers several candidate cerebrospinal fluid (CSF) biomarkers for PD based on the recent literature and meta-analysis data. The decrease of α-synuclein in PD is supported by meta-analyses with modest reproducibility, and a decrease of amyloid β42 is seen as a prognostic marker for cognitive decline. Tau, phosphorylated tau (p-tau), and neurofilament light chains have been used to discriminate PD from other neurodegenerative disorders. This article also describes more hopeful biochemical markers, such as neurotransmitters, oxidative stress markers, and other candidate biomarkers.
Collapse
Affiliation(s)
- Takayuki Katayama
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa 070-8610, Japan; (K.T.); (O.Y.)
- Correspondence: ; Tel.: +81-166-24-3181; Fax: +81-166-24-1125
| | - Jun Sawada
- Department of Neurology, Asahikawa Medical University Hospital, Asahikawa 078-8510, Japan;
| | - Kae Takahashi
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa 070-8610, Japan; (K.T.); (O.Y.)
| | - Osamu Yahara
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa 070-8610, Japan; (K.T.); (O.Y.)
| |
Collapse
|
15
|
Jiang R, Rong C, Ke R, Meng S, Yan X, Ke H, Wu S. Differential proteomic analysis of serum exosomes reveals alterations in progression of Parkinson disease. Medicine (Baltimore) 2019; 98:e17478. [PMID: 31593110 PMCID: PMC6799836 DOI: 10.1097/md.0000000000017478] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Exosomes are nanometer-sized vesicles with intercellular communication functions, and their encapsulated proteins may participate in the pathological process of neurodegenerative disorders. The aim of this study was to identify the protein changes of serum exosomes in Parkinson disease (PD) patients with different disease progress types, and to identify potential biomarkers. The exosomes of PD patients with different severity and healthy control group were isolated from serum. The exosome proteins were analyzed by mass spectrometry with label-free quantitative proteomics. A total of 429 proteins were identified, of which 14 were significantly different in mild and severe PD patients. The expression levels of 7 proteins, including pigmented epithelium-derived factor, afamin, apolipoprotein D and J, were significantly increased in PD patients. The expression levels of 7 proteins, including complement C1q and protein Immunoglobulin Lambda Variable 1-33 (IGLV1-33)Cluster -33, were decreased in PD patients. These differentially expressed proteins were analyzed by gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis, which confirmed that the interaction between prion diseases and ECM receptors was the most significant pathways of enrichment. The changes of proteins and pathways may be related to the pathophysiological mechanism of PD. Therefore, some of these proteins could be considered as potential biomarkers for early PD diagnosis.
Collapse
Affiliation(s)
- Ruilai Jiang
- Department of Emergency, the Second People's Hospital of Lishui, Lishui, Zhejiang Province
| | - Chunjiao Rong
- Department of Emergency, the Second People's Hospital of Lishui, Lishui, Zhejiang Province
| | - Ronghu Ke
- Department of Plastic and Reconstructive Surgery
| | - Shuiyan Meng
- Department of Emergency, the Second People's Hospital of Lishui, Lishui, Zhejiang Province
| | - Xiumei Yan
- Department of Emergency, the Second People's Hospital of Lishui, Lishui, Zhejiang Province
| | - Honglin Ke
- Department of Emergency, Huashan Hospital, Fudan University School of Medicine, Shanghai, China
| | - Shaochang Wu
- Department of Emergency, the Second People's Hospital of Lishui, Lishui, Zhejiang Province
| |
Collapse
|
16
|
Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front Pharmacol 2019; 10:1008. [PMID: 31572186 PMCID: PMC6751310 DOI: 10.3389/fphar.2019.01008] [Citation(s) in RCA: 482] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative diseases share the fact that they derive from altered proteins that undergo an unfolding process followed by formation of β-structures and a pathological tendency to self-aggregate in neuronal cells. This is a characteristic of tau protein in Alzheimer’s disease and several tauopathies associated with tau unfolding, α-synuclein in Parkinson’s disease, and huntingtin in Huntington disease. Usually, the self-aggregation products are toxic to these cells, and toxicity spreads all over different brain areas. We have postulated that these protein unfolding events are the molecular alterations that trigger several neurodegenerative disorders. Most interestingly, these events occur as a result of neuroinflammatory cascades involving alterations in the cross-talks between glial cells and neurons as a consequence of the activation of microglia and astrocytes. The model we have hypothesized for Alzheimer’s disease involves damage signals that promote glial activation, followed by nuclear factor NF-kβ activation, synthesis, and release of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-6, and IL-12 that affect neuronal receptors with an overactivation of protein kinases. These patterns of pathological events can be applied to several neurodegenerative disorders. In this context, the involvement of innate immunity seems to be a major paradigm in the pathogenesis of these diseases. This is an important element for the search for potential therapeutic approaches for all these brain disorders.
Collapse
Affiliation(s)
- Leonardo Guzman-Martinez
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Ricardo B Maccioni
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Andrade
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Leonardo Patricio Navarrete
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - María Gabriela Pastor
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Nicolas Ramos-Escobar
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| |
Collapse
|
17
|
Vida C, Kobayashi H, Garrido A, Martínez de Toda I, Carro E, Molina JA, De la Fuente M. Lymphoproliferation Impairment and Oxidative Stress in Blood Cells from Early Parkinson's Disease Patients. Int J Mol Sci 2019; 20:ijms20030771. [PMID: 30759742 PMCID: PMC6386872 DOI: 10.3390/ijms20030771] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/28/2019] [Accepted: 02/02/2019] [Indexed: 12/20/2022] Open
Abstract
In Parkinson’s Disease (PD), the peripheral changes in the functional capacity and redox state of immune cells has been scarcely investigated, especially in the early PD stages. Aging is a risk factor for PD, and the age-related impairment of the immune system, based on a chronic-oxidative stress situation, is involved in the rate of aging. We analyzed several functions in isolated peripheral blood neutrophils and mononuclear cells from PD stage 2 patients, and compared the results to those in healthy elderly and adult controls. Several oxidative stress and damage parameters were studied in whole blood cells. The results showed an impairment of the lymphoproliferative response in stimulated conditions in the PD patients compared with age-matched controls, who also showed typical immunosenescence in comparison with adult individuals. Higher oxidative stress and damage were observed in whole blood cells from PD patients (lower glutathione peroxidase activity, and higher oxidized glutathione and malondialdehyde contents). Our results suggest an accelerated immunosenescence in PD stage 2, and that several of the parameters studied could be appropriate peripheral biomarkers in the early stages of PD.
Collapse
Affiliation(s)
- Carmen Vida
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
| | - Hikaru Kobayashi
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
| | - Antonio Garrido
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
| | - Irene Martínez de Toda
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
| | - Eva Carro
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain.
| | - José Antonio Molina
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain.
| | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain.
- Institute of Biomedical Research Hospital 12 Octubre (imas12), 28041 Madrid, Spain.
| |
Collapse
|
18
|
Gupta AK, Rani K, Swarnkar S, Kumar GK, Khan MI, Pokhriyal R, Kumar DR, Goyal V, Tripathi M, Gupta R, Chadda RK, Vanamail P, Hariprasad G. Evaluation of Serum Apolipoprotein E as a Potential Biomarker for Pharmacological Therapeutic Efficacy Monitoring in Dopamine Dictated Disease Spectrum of Schizophrenia and Parkinson's disease: A Preliminary Study. J Cent Nerv Syst Dis 2018; 10:1179573518803585. [PMID: 30327579 PMCID: PMC6178121 DOI: 10.1177/1179573518803585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/02/2018] [Indexed: 11/15/2022] Open
Abstract
AIM OF THE STUDY Parkinson's disease and schizophrenia are disease end points of dopaminergic deficit and hyperactivity, respectively, in the mid brain. Accordingly, current medications aim to restore normal dopamine levels, overshooting of which results in adverse effects of psychosis and extra-pyramidal symptoms, respectively. There are currently no available laboratory tests to guide treatment decisions or help predict adverse side effects of the drugs. The aim was to therefore explore the possibility of using apolipoprotein E as a biomarker to monitor pharmacological intervention in dopamine dictated states of Parkinson's disease and schizophrenia for optimum therapy. METHODS Naïve and treated, Parkinson's disease and schizophrenic patients were recruited from neurology and psychiatry clinics. Serum of healthy volunteers was collected as controls. Serum concentrations of apolipoprotein E was estimated by enzyme-linked immunosorbent assay (ELISA). Pathway analysis was carried out to delineate the interactions of apolipoprotein E in Parkinson's disease and schizophrenia. RESULTS Apolipoprotein E levels are higher in Parkinson's disease patients as compared with schizophrenic samples (P < .05). Also, post-treatment apolipoprotein E levels in both disease states were at par with levels seen in healthy controls. The interactions of apolipoprotein E validate the results and place the differential expression of the protein in Parkinson's disease and schizophrenia in the right perspective. CONCLUSION Apolipoprotein E concentration across the dopaminergic spectrum suggests that it can be pursued not only as a potential biomarker in schizophrenia and Parkinson's disease, but can also be an effective tool for clinicians to determine efficacy of drug-based therapy.
Collapse
Affiliation(s)
- Ashish Kumar Gupta
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Komal Rani
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Surabhi Swarnkar
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Gaurav Khunger Kumar
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Mohd Imran Khan
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Ruchika Pokhriyal
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Domada Ratna Kumar
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Vinay Goyal
- Department of Neurology, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Rishab Gupta
- Department of Psychiatry, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Rakesh Kumar Chadda
- Department of Psychiatry, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Perumal Vanamail
- Department of Biostatistics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| | - Gururao Hariprasad
- Department of Biophysics, All India
Institute of Medical Sciences, New Delhi, New Delhi, India
| |
Collapse
|
19
|
Li L, Liu MS, Li GQ, Tang J, Liao Y, Zheng Y, Guo TL, Kang X, Yuan MT. Relationship between Apolipoprotein Superfamily and Parkinson's Disease. Chin Med J (Engl) 2018; 130:2616-2623. [PMID: 29067960 PMCID: PMC5678263 DOI: 10.4103/0366-6999.217092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Parkinson's disease (PD) is featured with motor disorder and nonmotor manifestations including psychological symptoms, autonomic nervous system dysfunction, and paresthesia, which results in great inconvenience to the patients’ life. The apolipoprotein (Apo) superfamily, as a group of potentially modifiable biomarkers in clinical practice, is of increasing significance in the diagnosis, evaluation, and prognosis of PD. The present review summarized the current understanding and emerging findings of the relationship between Apo superfamily and PD. Data Sources: All literatures were identified by systematically searching PubMed, Embase, and Cochrane electronic databases with terms “Parkinson disease,” “apolipoprotein,” and their synonyms until May 2017. Study Selection: We have thoroughly examined titles and abstracts of all the literatures that met our search strategy and the full text if the research is identified or not so definite. Reference lists of retrieved articles were also scrutinized for additional relevant studies. Results: The levels of plasma ApoA1 are inversely correlated with the risk of PD and the lower levels of ApoA1 trend toward association with poorer motor performance. Higher ApoD expression in neurons represents more puissant protection against PD, which is critical in delaying the neurodegeneration process of PD. It is suggested that APOE alleles are related to development and progression of cognitive decline and age of PD onset, but conclusions are not completely identical, which may be attributed to different ApoE isoforms. APOJ gene expressions are upregulated in PD patients and it is possible that high ApoJ level is an indicator of PD dementia and correlates with specific phenotypic variations in PD. Conclusions: The Apo superfamily has been proved to be closely involved in the initiation, progression, and prognosis of PD. Apos and their genes are of great value in predicting the susceptibility of PD and hopeful to become the target of medical intervention to prevent the onset of PD or slow down the progress. Therefore, further large-scale studies are warranted to elucidate the precise mechanisms of Apos in PD.
Collapse
Affiliation(s)
- Lin Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Ming-Su Liu
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Guang-Qin Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jie Tang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yan Liao
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yang Zheng
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Tong-Li Guo
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xin Kang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Mao-Ting Yuan
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
20
|
Neuroimmune Tau Mechanisms: Their Role in the Progression of Neuronal Degeneration. Int J Mol Sci 2018; 19:ijms19040956. [PMID: 29570615 PMCID: PMC5979395 DOI: 10.3390/ijms19040956] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/15/2022] Open
Abstract
Progressive neurodegenerative pathologies in aged populations are an issue of major concern worldwide. The microtubule-associated protein tau is able to self-aggregate to form abnormal supramolecular structures that include small oligomers up to complex polymers. Tauopathies correspond to a group of diseases that share tau pathology as a common etiological agent. Since microglial cells play a preponderant role in innate immunity and are the main source of proinflammatory factors in the central nervous system (CNS), the alterations in the cross-talks between microglia and neuronal cells are the main focus of studies concerning the origins of tauopathies. According to evidence from a series of studies, these changes generate a feedback mechanism reactivating microglia and provoking constant cellular damage. Thus, the previously summarized mechanisms could explain the onset and progression of different tauopathies and their functional/behavioral effects, opening the window towards an understanding of the molecular basis of anomalous tau interactions. Despite clinical and pathological differences, increasing experimental evidence indicates an overlap between tauopathies and synucleinopathies, considering that neuroinflammatory events are involved and the existence of protein misfolding. Neurofibrillary tangles of pathological tau (NFT) and Lewy bodies appear to coexist in certain brain areas. Thus, the co-occurrence of synucleinopathies with tauopathies is evidenced by several investigations, in which NFT were found in the substantia nigra of patients with Parkinson’s disease, suggesting that the pathologies share some common features at the level of neuroinflammatory events.
Collapse
|
21
|
Mashayekhi S, Salehi Z, Zahiri Z, Mirzajani E, Shahangian S. Correlation between serum and peritoneal fluid glutathione S-transferases T1 concentration with different stages of endometriosis. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2018. [DOI: 10.1016/j.mefs.2017.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
22
|
Chiu CC, Yeh TH, Lai SC, Weng YH, Huang YC, Cheng YC, Chen RS, Huang YZ, Hung J, Chen CC, Lin WY, Chang HC, Chen YJ, Chen CL, Chen HY, Lin YW, Wu-Chou YH, Wang HL, Lu CS. Increased Rab35 expression is a potential biomarker and implicated in the pathogenesis of Parkinson's disease. Oncotarget 2018; 7:54215-54227. [PMID: 27509057 PMCID: PMC5342336 DOI: 10.18632/oncotarget.11090] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/29/2016] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is the second common neurodegenerative disease. Identification of biomarkers for early diagnosis and prediction of disease progression is important. The present comparative proteomic study of serum samples using two-dimensional fluorescence differential gel electrophoresis followed by ELISA confirmation demonstrated that protein expression of Rab35 was increased in PD patients compared with matched control subjects and other parkinsonian disorders, progressive supranuclear palsy (PSP) and multiple system atrophy (MSA). The serum level of Rab35 was significantly correlated with the age at onset of PD. The median age of onset in patients with higher Rab35 serum level was 5 years younger than those with lower Rab35 serum level. There was a positive correlation between the Rab35 level and disease duration of PD. Moreover, the protein expression of Rab35 was increased in the substantia nigra but not in the striatum of mouse models of PD, including MPTP-treated mice, rotenone-treated mice, (R1441C) LRRK2 or (G2019S) LRRK2 transgenic mice. Furthermore, overexpression of Rab35 increased the aggregation and secretion of mutant A53T α-synuclein in dopaminergic SH-SY5Y cells. Co-expression of Rab35 with wild-type or A53T α-synuclein in SH-SY5Y cells deteriorated cell death. Our results suggest that Rab35 is potentially useful in the differential diagnosis of parkinsonian disorders and is implicated in the pathogenesis of PD.
Collapse
Affiliation(s)
- Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Szu-Chia Lai
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsin Weng
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yi-Chuan Cheng
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Zu Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Cognitive Neuroscience, National Central University, Taoyuan,Taiwan
| | - June Hung
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiung-Chu Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wey-Yil Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chen Chang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Yu-Jie Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chao-Lang Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hsin-Yi Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yan-Wei Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yah-Huei Wu-Chou
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Department of Physiology and Pharmacology, Chang Gung University School of Medicine, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University School of Medicine, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
23
|
Why should neuroscientists worry about iron? The emerging role of ferroptosis in the pathophysiology of neuroprogressive diseases. Behav Brain Res 2017; 341:154-175. [PMID: 29289598 DOI: 10.1016/j.bbr.2017.12.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a unique form of programmed death, characterised by cytosolic accumulation of iron, lipid hydroperoxides and their metabolites, and effected by the fatal peroxidation of polyunsaturated fatty acids in the plasma membrane. It is a major driver of cell death in neurodegenerative neurological diseases. Moreover, cascades underpinning ferroptosis could be active drivers of neuropathology in major psychiatric disorders. Oxidative and nitrosative stress can adversely affect mechanisms and proteins governing cellular iron homeostasis, such as the iron regulatory protein/iron response element system, and can ultimately be a source of abnormally high levels of iron and a source of lethal levels of lipid membrane peroxidation. Furthermore, neuroinflammation leads to the upregulation of divalent metal transporter1 on the surface of astrocytes, microglia and neurones, making them highly sensitive to iron overload in the presence of high levels of non-transferrin-bound iron, thereby affording such levels a dominant role in respect of the induction of iron-mediated neuropathology. Mechanisms governing systemic and cellular iron homeostasis, and the related roles of ferritin and mitochondria are detailed, as are mechanisms explaining the negative regulation of ferroptosis by glutathione, glutathione peroxidase 4, the cysteine/glutamate antiporter system, heat shock protein 27 and nuclear factor erythroid 2-related factor 2. The potential role of DJ-1 inactivation in the precipitation of ferroptosis and the assessment of lipid peroxidation are described. Finally, a rational approach to therapy is considered, with a discussion on the roles of coenzyme Q10, iron chelation therapy, in the form of deferiprone, deferoxamine (desferrioxamine) and deferasirox, and N-acetylcysteine.
Collapse
|
24
|
Mortiboys H, Macdonald R, Payne T, Sassani M, Jenkins T, Bandmann O. Translational approaches to restoring mitochondrial function in Parkinson's disease. FEBS Lett 2017; 592:776-792. [PMID: 29178330 DOI: 10.1002/1873-3468.12920] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/04/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022]
Abstract
There is strong evidence of a key role for mitochondrial dysfunction in both sporadic and all forms of familial Parkinson's disease (PD). However, none of the clinical trials carried out with putative mitochondrial rescue agents have been successful. Firm establishment of a wet biomarker or a reliable readout from imaging studies detecting mitochondrial dysfunction and reflecting disease progression is also awaited. We will provide an overview of our current knowledge about mitochondrial dysfunction in PD and related drug screens. We will also summarise previously undertaken mitochondrial wet biomarker studies and relevant imaging studies with particular focus on 31P-MRI spectroscopy. We will conclude with an overview of clinical trials which tested putative mitochondrial rescue agents in PD patients.
Collapse
Affiliation(s)
- Heather Mortiboys
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Ruby Macdonald
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Thomas Payne
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Matilde Sassani
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Thomas Jenkins
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| | - Oliver Bandmann
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK
| |
Collapse
|
25
|
Connor DE, Chaitanya GV, Chittiboina P, McCarthy P, Scott LK, Schrott L, Minagar A, Nanda A, Alexander JS. Variations in the cerebrospinal fluid proteome following traumatic brain injury and subarachnoid hemorrhage. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2017; 24:169-183. [PMID: 28549769 PMCID: PMC7303909 DOI: 10.1016/j.pathophys.2017.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Proteomic analysis of cerebrospinal fluid (CSF) has shown great promise in identifying potential markers of injury in neurodegenerative diseases [1-13]. Here we compared CSF proteomes in healthy individuals, with patients diagnosed with traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) in order to characterize molecular biomarkers which might identify these different clinical states and describe different molecular mechanisms active in each disease state. METHODS Patients presenting to the Neurosurgery service at the Louisiana State University Hospital-Shreveport with an admitting diagnosis of TBI or SAH were prospectively enrolled. Patients undergoing CSF sampling for diagnostic procedures were also enrolled as controls. CSF aliquots were subjected to 2-dimensional gel electrophoresis (2D GE) and spot percentage densities analyzed. Increased or decreased spot expression (compared to controls) was defined in terms of in spot percentages, with spots showing consistent expression change across TBI or SAH specimens being followed up by Matrix-Assisted Laser Desorption/Ionization mass spectrometry (MALDI-MS). Polypeptide masses generated were matched to known standards using a search of the NCBI and/or GenPept databases for protein matches. Eight hundred fifteen separately identifiable polypeptide migration spots were identified on 2D GE gels. MALDI-MS successfully identified 13 of 22 selected 2D GE spots as recognizable polypeptides. RESULTS Statistically significant changes were noted in the expression of fibrinogen, carbonic anhydrase-I (CA-I), peroxiredoxin-2 (Prx-2), both α and β chains of hemoglobin, serotransferrin (Tf) and N-terminal haptoglobin (Hp) in TBI and SAH specimens, as compared to controls. The greatest mean fold change among all specimens was seen in CA-I and Hp at 30.7 and -25.7, respectively. TBI specimens trended toward greater mean increases in CA-I and Prx-2 and greater mean decreases in Hp and Tf. CONCLUSIONS Consistent CSF elevation of CA-I and Prx-2 with concurrent depletion of Hp and Tf may represent a useful combination of biomarkers for the prediction of severity and prognosis following brain injury.
Collapse
Affiliation(s)
- David E Connor
- Baptist Health Neurosurgery Arkansas, Little Rock, AR, United States.
| | - Ganta V Chaitanya
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD, United States.
| | - Paul McCarthy
- Department of Medicine, Sect. of Nephrology, University of Maryland, Baltimore, MD, United States.
| | - L Keith Scott
- Department of Critical Care Medicine, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Lisa Schrott
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - Anil Nanda
- Department of Neurosurgery, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, LA, United States.
| |
Collapse
|
26
|
Andersen AD, Binzer M, Stenager E, Gramsbergen JB. Cerebrospinal fluid biomarkers for Parkinson's disease - a systematic review. Acta Neurol Scand 2017; 135:34-56. [PMID: 26991855 DOI: 10.1111/ane.12590] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2016] [Indexed: 12/14/2022]
Abstract
Diagnosis of Parkinson's disease (PD) relies on clinical history and physical examination, but misdiagnosis is common in early stages. Identification of biomarkers for PD may allow early and more precise diagnosis and monitoring of dopamine replacement strategies and disease modifying treatments. Developments in analytical chemistry allow the detection of large numbers of molecules in plasma or cerebrospinal fluid, associated with the pathophysiology or pathogenesis of PD. This systematic review includes cerebrospinal fluid biomarker studies focusing on different disease pathways: oxidative stress, neuroinflammation, lysosomal dysfunction and proteins involved in PD and other neurodegenerative disorders, focusing on four clinical domains: their ability to (1) distinguish PD from healthy subjects and other neurodegenerative disorders as well as their relation to (2) disease duration after initial diagnosis, (3) severity of disease (motor symptoms) and (4) cognitive dysfunction. Oligomeric alpha-synuclein might be helpful in the separation of PD from controls. Through metabolomics, changes in purine and tryptophan metabolism have been discovered in patients with PD. Neurofilament light chain (NfL) has a significant role in distinguishing PD from other neurodegenerative diseases. Several oxidative stress markers are related to disease severity, with the antioxidant urate also having a prognostic value in terms of disease severity. Increased levels of amyloid and tau-proteins correlate with cognitive decline and may have prognostic value for cognitive deficits in PD. In the future, larger longitudinal studies, corroborating previous research on viable biomarker candidates or using metabolomics identifying a vast amount of potential biomarkers, could be a good approach.
Collapse
Affiliation(s)
- A. D. Andersen
- Department of Neurology; Hospital of Southern Jutland; Sønderborg Denmark
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
| | - M. Binzer
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
| | - E. Stenager
- Institute of Regional Health Research; University of Southern Denmark; Odense Denmark
- Focused Research Group in Neurology; Hospital of Southern Jutland; Sønderborg Denmark
- The Multiple Sclerosis Clinic of Southern Jutland; (Vejle, Sonderborg, Esbjerg) Denmark
| | - J. B. Gramsbergen
- Institute of Molecular Medicine, Neurobiological Research; University of Southern Denmark; Odense Denmark
| |
Collapse
|
27
|
Dominiak A, Wilkaniec A, Wroczyński P, Adamczyk A. Selenium in the Therapy of Neurological Diseases. Where is it Going? Curr Neuropharmacol 2016; 14:282-99. [PMID: 26549649 PMCID: PMC4857624 DOI: 10.2174/1570159x14666151223100011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 08/20/2015] [Accepted: 09/16/2015] [Indexed: 12/19/2022] Open
Abstract
Selenium (34Se), an antioxidant trace element, is an important regulator of brain function. These beneficial properties that Se possesses are attributed to its ability to be incorporated into selenoproteins as an amino acid. Several selenoproteins are expressed in the brain, in which some of them, e.g. glutathione peroxidases (GPxs), thioredoxin reductases (TrxRs) or selenoprotein P (SelP), are strongly involved in antioxidant defence and in maintaining intercellular reducing conditions. Since increased oxidative stress has been implicated in neurological disorders, including Parkinson’s disease, Alzheimer’s disease, stroke, epilepsy and others, a growing body of evidence suggests that Se depletion followed by decreased activity of Se-dependent enzymes may be important factors connected with those pathologies. Undoubtedly, the remarkable progress that has been made in understanding the biological function of Se in the brain has opened up new potential possibilities for the treatment of neurological diseases by using Se as a potential drug. However, further research in the search for optimal Se donors is necessary in order to achieve an effective and safe therapeutic income.
Collapse
Affiliation(s)
| | - Anna Wilkaniec
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland.
| | | | | |
Collapse
|
28
|
Yang P, Pavlovic D, Waldvogel H, Dragunow M, Synek B, Turner C, Faull R, Guan J. String Vessel Formation is Increased in the Brain of Parkinson Disease. JOURNAL OF PARKINSONS DISEASE 2016; 5:821-36. [PMID: 26444086 DOI: 10.3233/jpd-140454] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND String vessels are collapsed basement membrane without endothelium and have no function in circulation. String vessel formation contributes to vascular degeneration in Alzheimer disease. By comparing to age-matched control cases we have recently reported endothelial degeneration in brain capillaries of human Parkinson disease (PD). OBJECTIVE Current study evaluated changes of basement membrane of capillaries, string vessel formation and their association with astrocytes, blood-brain-barrier integrity and neuronal degeneration in PD. METHODS Brain tissue from human cases of PD and age-matched controls was used. Immunohistochemical staining for collagen IV, GFAP, NeuN, tyrosine hydroxylase, fibrinogen and Factor VIII was evaluated by image analysis in the substantia nigra, caudate nucleus and middle frontal gyrus. RESULTS While the basement-membrane-associated vessel density was similar between the two groups, the density of string vessels was significantly increased in the PD cases, particularly in the substantia nigra. Neuronal degeneration was found in all brain regions. Astrocytes and fibrinogen were increased in the caudate nuclei of PD cases compared with control cases. CONCLUSIONS Endothelial degeneration and preservation of basement membrane result in an increase of string vessel formation in PD. The data may suggest a possible role for cerebral hypoperfusion in the neuronal degeneration characteristic of PD, which needs further investigation. Elevated astrocytosis in the caudate nucleus of PD cases could be associated with disruption of the blood-brain barrier in this brain region.
Collapse
Affiliation(s)
- Panzao Yang
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Darja Pavlovic
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Henry Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Beth Synek
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital Auckland, New Zealand
| | - Richard Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Jian Guan
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| |
Collapse
|
29
|
Halbgebauer S, Öckl P, Wirth K, Steinacker P, Otto M. Protein biomarkers in Parkinson's disease: Focus on cerebrospinal fluid markers and synaptic proteins. Mov Disord 2016; 31:848-60. [PMID: 27134134 DOI: 10.1002/mds.26635] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/06/2016] [Accepted: 03/09/2016] [Indexed: 01/06/2023] Open
Abstract
Despite extensive research, to date, no validated biomarkers for PD have been found. This review seeks to summarize studies approaching the detection of biomarker candidates for PD and introduce promising ones in more detail, with special attention to synaptic proteins. To this end, we performed a PubMed search and included studies using proteomic tools (2-dimensional difference in gel electrophoresis and/or mass spectrometry) for the comparison of samples from PD and control patients. We found 27 studies reporting more than 500 differentially expressed proteins in which a total of 28 were detected in 2 and 17 in 3 or more independent studies, including posttranslationally modified proteins. In addition, of these 500 proteins, 25 were found to be brain specific, and 14 were enriched in synapses. Special attention was given to the applicability of the biomarker regarding sampling procedures, that is, using CSF/serum material for diagnosis. Furthermore, presynaptic proteins involved in vesicle membrane fusion seem to be interesting candidates for future analyses. Nonetheless, even though such promising biomarker candidates for PD exist, validation of these biomarkers in large-scale clinical studies is necessary to evaluate the diagnostic potential. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Patrick Öckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
30
|
Gene dysregulation is restored in the Parkinson's disease MPTP neurotoxic mice model upon treatment of the therapeutic drug Cu(II)(atsm). Sci Rep 2016; 6:22398. [PMID: 26928495 PMCID: PMC4772163 DOI: 10.1038/srep22398] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/15/2016] [Indexed: 12/04/2022] Open
Abstract
The administration of MPTP selectively targets the dopaminergic system resulting in Parkinsonism-like symptoms and is commonly used as a mice model of Parkinson’s disease. We previously demonstrated that the neuroprotective compound CuII(atsm) rescues nigral cell loss and improves dopamine metabolism in the MPTP model. The mechanism of action of CuII(atsm) needs to be further defined to understand how the compound promotes neuronal survival. Whole genome transcriptomic profiling has become a popular method to examine the relationship between gene expression and function. Substantia nigra samples from MPTP-lesioned mice were evaluated using whole transcriptome sequencing to investigate the genes altered upon CuII(atsm) treatment. We identified 143 genes affected by MPTP lesioning that are associated with biological processes related to brain and cognitive development, dopamine synthesis and perturbed synaptic neurotransmission. Upon CuII(atsm) treatment, the expression of 40 genes involved in promoting dopamine synthesis, calcium signaling and synaptic plasticity were restored which were validated by qRT-PCR. The study provides the first detailed whole transcriptomic analysis of pathways involved in MPTP-induced Parkinsonism. In addition, we identify key therapeutic pathways targeted by a potentially new class of neuroprotective agents which may provide therapeutic benefits for other neurodegenerative disorders.
Collapse
|
31
|
Přikrylová Vranová H, Hényková E, Mareš J, Kaiserová M, Menšíková K, Vaštík M, Hluštík P, Zapletalová J, Strnad M, Stejskal D, Kaňovský P. Clusterin CSF levels in differential diagnosis of neurodegenerative disorders. J Neurol Sci 2016; 361:117-21. [DOI: 10.1016/j.jns.2015.12.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 11/20/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022]
|
32
|
|
33
|
Martín de Pablos A, García-Moreno JM, Fernández E. Does the Cerebrospinal Fluid Reflect Altered Redox State But Not Neurotrophic Support Loss in Parkinson's Disease? Antioxid Redox Signal 2015; 23:893-8. [PMID: 26263410 DOI: 10.1089/ars.2015.6423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alteration in neurotrophic factors support and antioxidant defenses in the central nervous system (CNS) along with deficit of ferritin have been associated with idiopathic Parkinson's disease (PD). The objectives were to analyze in the cerebrospinal fluid (CSF) of patients with PD and controls the following: (i) the levels of the neuroprotectant factors glial cell line-derived neurotrophic factor, persephin, neurturin, and brain-derived neurotrophic factor, (ii) the levels of transforming growth factor-β1 (TGFβ1) and transforming growth factor-β2 (TGFβ2), proinflammatory factors, (iii) the activity of the antioxidant enzymes glutathione peroxidase (GPx), glutathione reductase (GR), glutathione-S-transferase (GST), catalase, superoxide dismutases (SODs), and peroxiredoxins (PRDxs), and (iv) ferritin levels. The study revealed that, among neurotrophic factors, only TGFβ1 levels were found to be enhanced in patients with PD (early, p < 0.05; advanced, p < 0.02). Regarding antioxidant enzymes, the activity of GPx, catalase, and PRDxs, all hydrogen peroxide scavengers, was found to be significantly reduced in patients (GPx, p < 0.001; catalase, p < 0.01; PRDxs, p < 0.01, one-way analysis of variance). Finally, ferritin content in CSF was significantly diminished over time in patients (early, p < 0.01, -49%; advanced, p < 0.001, -80.7%). Our observations lead to the hypothesis that parkinsonian patients suffer from a serious disturbance of redox state in the CNS, as evaluated through the CSF, characterized by reduced hydrogen peroxide scavenging and iron storage.
Collapse
Affiliation(s)
- Angel Martín de Pablos
- 1 Laboratory of Neurophysiology and Molecular Neurology (BIO127), Department of Medical Physiology and Biophysics, University of Seville , Seville, Spain .,2 Department of Surgery, University of Seville and Department of Anesthesiology, Hospital Macarena, Seville, Spain
| | - José-Manuel García-Moreno
- 1 Laboratory of Neurophysiology and Molecular Neurology (BIO127), Department of Medical Physiology and Biophysics, University of Seville , Seville, Spain .,3 Neuroscience Clinical Unit & Asociación Neuroinvest, Hospital Macarena , Seville, Spain
| | - Emilio Fernández
- 1 Laboratory of Neurophysiology and Molecular Neurology (BIO127), Department of Medical Physiology and Biophysics, University of Seville , Seville, Spain
| |
Collapse
|
34
|
Abstract
Human cerebrospinal fluid (CSF) contains diverse lipid particles, including lipoproteins that are distinct from their plasma counterparts and contain apolipoprotein (apo) E isoforms, apoJ, and apoAI, and extracellular vesicles, which can be detected by annexin V binding. The aim of this study was to develop a method to quantify CSF particles and evaluate their relationship to aging and neurodegenerative diseases. We used a flow cytometric assay to detect annexin V-, apoE-, apoAI-, apoJ-, and amyloid (A) β42-positive particles in CSF from 131 research volunteers who were neurologically normal or had mild cognitive impairment (MCI), Alzheimer disease (AD) dementia, or Parkinson disease. APOE ε4/ε4 participants had CSF apoE-positive particles that were more frequently larger but at an 88% lower level versus those in APOE ε3/ε3 or APOE ε3/ε4 patients; this finding was reproduced in conditioned medium from mouse primary glial cell cultures with targeted replacement of apoE. Cerebrospinal fluid apoE-positive and β-amyloid (Aβ42)-positive particle concentrations were persistently reduced one-third to one-half in middle and older age subjects; apoAI-positive particle concentration progressively increased approximately 2-fold with age. Both apoAI-positive and annexin V-positive CSF particle levels were reduced one-third to one-half in CSF of MCI and/or AD dementia patients versus age-matched controls. Our approach provides new methods to investigate CNS lipid biology in relation to neurodegeneration and perhaps develop new biomarkers for diagnosis or treatment monitoring.
Collapse
|
35
|
Shevchenko G, Konzer A, Musunuri S, Bergquist J. Neuroproteomics tools in clinical practice. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:705-17. [PMID: 25680928 DOI: 10.1016/j.bbapap.2015.01.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/30/2015] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are characterized by neuronal impairment that leads to disease-specific changes in the neuronal proteins. The early diagnosis of these disorders is difficult, thus, the need for identifying, developing and using valid clinically applicable biomarkers that meet the criteria of precision, specificity and repeatability is very vital. The application of rapidly emerging technology such as mass spectrometry (MS) in proteomics has opened new avenues to accelerate biomarker discovery, both for diagnostic as well as for prognostic purposes. This review summarizes the most recent advances in the mass spectrometry-based neuroproteomics and analyses the current and future directions in the biomarker discovery for the neurodegenerative diseases. This article is part of a Special Issue entitled: Neuroproteomics: Applications in Neuroscience and Neurology.
Collapse
Affiliation(s)
- Ganna Shevchenko
- Analytical Chemistry, Department of Chemistry-BMC and SciLife Lab, Uppsala University, 75124 Uppsala, Sweden
| | - Anne Konzer
- Analytical Chemistry, Department of Chemistry-BMC and SciLife Lab, Uppsala University, 75124 Uppsala, Sweden
| | - Sravani Musunuri
- Analytical Chemistry, Department of Chemistry-BMC and SciLife Lab, Uppsala University, 75124 Uppsala, Sweden
| | - Jonas Bergquist
- Analytical Chemistry, Department of Chemistry-BMC and SciLife Lab, Uppsala University, 75124 Uppsala, Sweden.
| |
Collapse
|
36
|
Mazzetti AP, Fiorile MC, Primavera A, Lo Bello M. Glutathione transferases and neurodegenerative diseases. Neurochem Int 2015; 82:10-8. [PMID: 25661512 DOI: 10.1016/j.neuint.2015.01.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/23/2015] [Accepted: 01/27/2015] [Indexed: 02/08/2023]
Abstract
There is substantial agreement that the unbalance between oxidant and antioxidant species may affect the onset and/or the course of a number of common diseases including Parkinson's and Alzheimer's diseases. Many studies suggest a crucial role for oxidative stress in the first phase of aging, or in the pathogenesis of various diseases including neurological ones. Particularly, the role exerted by glutathione and glutathione-related enzymes (Glutathione Transferases) in the nervous system appears more relevant, this latter tissue being much more vulnerable to toxins and oxidative stress than other tissues such as liver, kidney or muscle. The present review addresses the question by focusing on the results obtained by specimens from patients or by in vitro studies using cells or animal models related to Parkinson's and Alzheimer's diseases. In general, there is an association between glutathione depletion and Parkinson's or Alzheimer's disease. In addition, a significant decrease of glutathione transferase activity in selected areas of brain and in ventricular cerebrospinal fluid was found. For some glutathione transferase genes there is also a correlation between polymorphisms and onset/outcome of neurodegenerative diseases. Thus, there is a general agreement about the protective effect exerted by glutathione and glutathione transferases but no clear answer about the mechanisms underlying this crucial role in the insurgence of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Mario Lo Bello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
37
|
Beach TG, Adler CH, Sue LI, Serrano G, Shill HA, Walker DG, Lue L, Roher AE, Dugger BN, Maarouf C, Birdsill AC, Intorcia A, Saxon-Labelle M, Pullen J, Scroggins A, Filon J, Scott S, Hoffman B, Garcia A, Caviness JN, Hentz JG, Driver-Dunckley E, Jacobson SA, Davis KJ, Belden CM, Long KE, Malek-Ahmadi M, Powell JJ, Gale LD, Nicholson LR, Caselli RJ, Woodruff BK, Rapscak SZ, Ahern GL, Shi J, Burke AD, Reiman EM, Sabbagh MN. Arizona Study of Aging and Neurodegenerative Disorders and Brain and Body Donation Program. Neuropathology 2015; 35:354-89. [PMID: 25619230 DOI: 10.1111/neup.12189] [Citation(s) in RCA: 356] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/11/2014] [Indexed: 12/13/2022]
Abstract
The Brain and Body Donation Program (BBDP) at Banner Sun Health Research Institute (http://www.brainandbodydonationprogram.org) started in 1987 with brain-only donations and currently has banked more than 1600 brains. More than 430 whole-body donations have been received since this service was commenced in 2005. The collective academic output of the BBDP is now described as the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND). Most BBDP subjects are enrolled as cognitively normal volunteers residing in the retirement communities of metropolitan Phoenix, Arizona. Specific recruitment efforts are also directed at subjects with Alzheimer's disease, Parkinson's disease and cancer. The median age at death is 82. Subjects receive standardized general medical, neurological, neuropsychological and movement disorders assessments during life and more than 90% receive full pathological examinations by medically licensed pathologists after death. The Program has been funded through a combination of internal, federal and state of Arizona grants as well as user fees and pharmaceutical industry collaborations. Subsets of the Program are utilized by the US National Institute on Aging Arizona Alzheimer's Disease Core Center and the US National Institute of Neurological Disorders and Stroke National Brain and Tissue Resource for Parkinson's Disease and Related Disorders. Substantial funding has also been received from the Michael J. Fox Foundation for Parkinson's Research. The Program has made rapid autopsy a priority, with a 3.0-hour median post-mortem interval for the entire collection. The median RNA Integrity Number (RIN) for frozen brain and body tissue is 8.9 and 7.4, respectively. More than 2500 tissue requests have been served and currently about 200 are served annually. These requests have been made by more than 400 investigators located in 32 US states and 15 countries. Tissue from the BBDP has contributed to more than 350 publications and more than 200 grant-funded projects.
Collapse
Affiliation(s)
- Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Geidy Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Holly A Shill
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - LihFen Lue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Alex E Roher
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Chera Maarouf
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Alex C Birdsill
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Joel Pullen
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Jessica Filon
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Sarah Scott
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Angelica Garcia
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | | | | | - Kathryn J Davis
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Kathy E Long
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Lisa D Gale
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | | | | | | | - Jiong Shi
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Anna D Burke
- Banner Alzheimer Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
38
|
Licker V, Burkhard PR. Proteomics as a new paradigm to tackle Parkinson’s disease research challenges. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2014.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
39
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Agúndez JAG. Cerebrospinal fluid biochemical studies in patients with Parkinson's disease: toward a potential search for biomarkers for this disease. Front Cell Neurosci 2014; 8:369. [PMID: 25426023 PMCID: PMC4227512 DOI: 10.3389/fncel.2014.00369] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/20/2014] [Indexed: 12/14/2022] Open
Abstract
The blood-brain barrier supplies brain tissues with nutrients and filters certain compounds from the brain back to the bloodstream. In several neurodegenerative diseases, including Parkinson's disease (PD), there are disruptions of the blood-brain barrier. Cerebrospinal fluid (CSF) has been widely investigated in PD and in other parkinsonian syndromes with the aim of establishing useful biomarkers for an accurate differential diagnosis among these syndromes. This review article summarizes the studies reported on CSF levels of many potential biomarkers of PD. The most consistent findings are: (a) the possible role of CSF urate on the progression of the disease; (b) the possible relations of CSF total tau and phosphotau protein with the progression of PD and with the preservation of cognitive function in PD patients; (c) the possible value of CSF beta-amyloid 1-42 as a useful marker of further cognitive decline in PD patients, and (d) the potential usefulness of CSF neurofilament (NFL) protein levels in the differential diagnosis between PD and other parkinsonian syndromes. Future multicentric, longitudinal, prospective studies with long-term follow-up and neuropathological confirmation would be useful in establishing appropriate biomarkers for PD.
Collapse
Affiliation(s)
| | | | - Elena García-Martín
- Department of Biochemistry and Molecular Biology, University of ExtremaduraCáceres, Spain
- AMGenomicsCáceres, Spain
| | - José A. G. Agúndez
- AMGenomicsCáceres, Spain
- Department of Pharmacology, University of ExtremaduraCáceres, Spain
| |
Collapse
|
40
|
Swanson CR, Li K, Unger TL, Gallagher MD, Van Deerlin VM, Agarwal P, Leverenz J, Roberts J, Samii A, Gross RG, Hurtig H, Rick J, Weintraub D, Trojanowski JQ, Zabetian C, Chen-Plotkin AS. Lower plasma apolipoprotein A1 levels are found in Parkinson's disease and associate with apolipoprotein A1 genotype. Mov Disord 2014; 30:805-12. [PMID: 25227208 DOI: 10.1002/mds.26022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/25/2014] [Accepted: 07/16/2014] [Indexed: 01/17/2023] Open
Abstract
The discovery of novel plasma-based biomarkers could lead to new approaches in the treatment of Parkinson's disease (PD). Here, we explore the role of plasma apolipoprotein A1 (ApoA1) as a risk marker for PD and evaluate the influence of APOA1 promoter variation on plasma ApoA1 levels. Plasma ApoA1 and the single-nucleotide polymorphism, rs670, were assayed in a discovery cohort (cohort 1) of 301 PD patients, 80 normal controls (NCs), and 165 subjects with other neurodegenerative diseases, as well as a cohort (cohort 2) of 158 PD patients from a second clinical site. Additionally, rs670 was genotyped in a third cohort of 1,494 PD and 925 NC subjects from both clinical sites. Compared to both normal and disease controls, PD patients have lower plasma ApoA1 (P < 0.001 for both comparisons). Moreover, in PD patients, plasma ApoA1 levels are correlated with genotype at the APOA1 promoter polymorphism, rs670. Specifically, lower plasma ApoA1 levels were found in rs670 major allele (G) homozygotes in both cohort 1 (P = 0.009) and in a replication cohort (cohort 2; n = 158 PD patients; P = 0.024). Finally, evaluating rs670 genotype frequencies in 1,930 PD cases versus 997 NCs, the rs670 GG genotype shows a trend toward association (odds ratio: 1.1; P = 0.10) with PD. Our results are compatible with a model whereby circulating ApoA1 levels may be useful in risk-stratifying subjects for the development of PD, with higher ApoA1 levels suggesting relative protection. Future studies evaluating modulation of ApoA1 as a novel therapeutic strategy in PD are warranted. © 2014 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Christine R Swanson
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine Li
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Travis L Unger
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael D Gallagher
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Cellular and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pinky Agarwal
- Booth Gardner Parkinson's Care Center, Evergreen Hospital Medical Center, Kirkland, Washington, USA
| | - James Leverenz
- Cleveland Clinic Ruvo Center for Brain Health, Cleveland, Ohio, USA
| | - John Roberts
- Virginia Mason Medical Center, Seattle, Washington, USA
| | - Ali Samii
- VA Puget Sound Health Care System and Department of Neurology, University of Washington Seattle, Seattle, Washington, USA
| | - Rachel Goldmann Gross
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Howard Hurtig
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacqueline Rick
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cyrus Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington Seattle, Seattle, Washington, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Vranová HP, Hényková E, Kaiserová M, Menšíková K, Vaštík M, Mareš J, Hluštík P, Zapletalová J, Strnad M, Stejskal D, Kaňovský P. Tau protein, beta-amyloid1–42 and clusterin CSF levels in the differential diagnosis of Parkinsonian syndrome with dementia. J Neurol Sci 2014; 343:120-4. [DOI: 10.1016/j.jns.2014.05.052] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 05/20/2014] [Accepted: 05/22/2014] [Indexed: 10/25/2022]
|
42
|
Percy AJ, Yang J, Chambers AG, Simon R, Hardie DB, Borchers CH. Multiplexed MRM with Internal Standards for Cerebrospinal Fluid Candidate Protein Biomarker Quantitation. J Proteome Res 2014; 13:3733-3747. [DOI: 10.1021/pr500317d] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Andrew J. Percy
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
| | - Juncong Yang
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
| | - Andrew G. Chambers
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
| | - Romain Simon
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
| | - Darryl B. Hardie
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
| | - Christoph H. Borchers
- University of
Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, BC V8Z
7X8, Canada
- Department
of Biochemistry and Microbiology, University of Victoria, Petch Building
Room 207, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
43
|
Burgos K, Malenica I, Metpally R, Courtright A, Rakela B, Beach T, Shill H, Adler C, Sabbagh M, Villa S, Tembe W, Craig D, Van Keuren-Jensen K. Profiles of extracellular miRNA in cerebrospinal fluid and serum from patients with Alzheimer's and Parkinson's diseases correlate with disease status and features of pathology. PLoS One 2014; 9:e94839. [PMID: 24797360 PMCID: PMC4010405 DOI: 10.1371/journal.pone.0094839] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 02/13/2014] [Indexed: 01/09/2023] Open
Abstract
The discovery and reliable detection of markers for neurodegenerative diseases have been complicated by the inaccessibility of the diseased tissue--such as the inability to biopsy or test tissue from the central nervous system directly. RNAs originating from hard to access tissues, such as neurons within the brain and spinal cord, have the potential to get to the periphery where they can be detected non-invasively. The formation and extracellular release of microvesicles and RNA binding proteins have been found to carry RNA from cells of the central nervous system to the periphery and protect the RNA from degradation. Extracellular miRNAs detectable in peripheral circulation can provide information about cellular changes associated with human health and disease. In order to associate miRNA signals present in cell-free peripheral biofluids with neurodegenerative disease status of patients with Alzheimer's and Parkinson's diseases, we assessed the miRNA content in cerebrospinal fluid and serum from postmortem subjects with full neuropathology evaluations. We profiled the miRNA content from 69 patients with Alzheimer's disease, 67 with Parkinson's disease and 78 neurologically normal controls using next generation small RNA sequencing (NGS). We report the average abundance of each detected miRNA in cerebrospinal fluid and in serum and describe 13 novel miRNAs that were identified. We correlated changes in miRNA expression with aspects of disease severity such as Braak stage, dementia status, plaque and tangle densities, and the presence and severity of Lewy body pathology. Many of the differentially expressed miRNAs detected in peripheral cell-free cerebrospinal fluid and serum were previously reported in the literature to be deregulated in brain tissue from patients with neurodegenerative disease. These data indicate that extracellular miRNAs detectable in the cerebrospinal fluid and serum are reflective of cell-based changes in pathology and can be used to assess disease progression and therapeutic efficacy.
Collapse
Affiliation(s)
- Kasandra Burgos
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Ivana Malenica
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Raghu Metpally
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Amanda Courtright
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Benjamin Rakela
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Thomas Beach
- Neurology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Holly Shill
- Neurology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Charles Adler
- Neurology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Marwan Sabbagh
- Neurology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Stephen Villa
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Waibhav Tembe
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - David Craig
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Kendall Van Keuren-Jensen
- Neurogenomics, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- * E-mail:
| |
Collapse
|
44
|
Lu W, Wan X, Liu B, Rong X, Zhu L, Li P, Li J, Wang L, Cui L, Wang X. Specific changes of serum proteins in Parkinson's disease patients. PLoS One 2014; 9:e95684. [PMID: 24769800 PMCID: PMC4000217 DOI: 10.1371/journal.pone.0095684] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 03/28/2014] [Indexed: 12/12/2022] Open
Abstract
The aim of this study is to identify and validate protein change in the serum from PD patients. We used serum samples from 21 PD patients and 20 age-matched normal people as control to conduct a comparative proteomic study. We performed 2-DE and analyzed the differentially expressed protein spots by LC-MS/MS. In PD group 13 spots were shown to be differentially expressed compared to control group. They were identified as 6 proteins. Among these, 3 proteins were confirmed by Western blot analysis. It showed that the frequency of fibrinogen γ-chain (FGG) appeared 70% in PD, which could not be detected in control group. The protein of inter-alpha-trypsin inhibitor heavy chain H4 (ITI-H4) was found to exist two forms in serum. The full size (120 kDa) of the protein was increased and the fragmented ITI-H4 (35 kDa) was decreased in PD group. The ratio of full size ITI-H4 to fragmented ITI-H4 in PD patients was 3.85±0.29-fold higher than in control group. Furthermore, fragmented Apo A-IV (∼26 kDa) was mainly detected in control group, while it was rare to be found in PD group. Above findings might be useful for diagnosis of PD. When the expressions of FGG and 120 kDa ITI-H4 are increase, as well as ∼26 kDa Apo A-IV disappear would provide strong evidence for PD.
Collapse
Affiliation(s)
- Wenwen Lu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinhua Wan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bin Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianfang Rong
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiang Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (LC); (XW)
| | - Xiaoliang Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (LC); (XW)
| |
Collapse
|
45
|
Adler CH, Dugger BN, Hinni ML, Lott DG, Driver-Dunckley E, Hidalgo J, Henry-Watson J, Serrano G, Sue LI, Nagel T, Duffy A, Shill HA, Akiyama H, Walker DG, Beach TG. Submandibular gland needle biopsy for the diagnosis of Parkinson disease. Neurology 2014; 82:858-64. [PMID: 24500652 DOI: 10.1212/wnl.0000000000000204] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE This study investigates salivary gland biopsies in living patients with Parkinson disease (PD). METHODS Patients with PD for ≥5 years underwent outpatient transcutaneous needle core biopsies (18-gauge or 16-gauge) of 1 submandibular gland. Minor salivary glands were removed via a small incision in the lower lip. Tissue was fixed in formalin and serial 6-µm paraffin sections were immunohistochemically stained for phosphorylated α-synuclein and reviewed for evidence of Lewy type α-synucleinopathy (LTS). RESULTS Fifteen patients with PD were biopsied: 9 female/6 male, mean age 68.7 years, mean PD duration 11.8 years. Twelve of the needle core biopsies had microscopically evident submandibular gland tissue to assess and 9/12 (75%) had LTS. Only 1/15 (6.7%) minor salivary gland biopsies were positive for LTS. Five patients had an adverse event; all were minor and transient. CONCLUSIONS This study demonstrates the feasibility of performing needle core biopsies of the submandibular gland in living patients with PD to assess LTS. Although this was a small study, this tissue biopsy method may be important for tissue confirmation of PD in patients being considered for invasive procedures and in research studies of other PD biomarkers.
Collapse
Affiliation(s)
- Charles H Adler
- From the Parkinson's Disease and Movement Disorders Center (C.H.A., E.D.-D., A.D.), Department of Neurology, Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (B.N.D., J.H., J.H.-W., G.S., L.I.S., H.A.S., D.G.W., T.G.B.), Sun City; the Department of Otolaryngology (M.L.H., D.G.L., T.N.), Mayo Clinic, Phoenix, AZ; and the Tokyo Institute of Psychiatry (H.A.), Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
van Dijk KD, Jongbloed W, Heijst JA, Teunissen CE, Groenewegen HJ, Berendse HW, van de Berg WD, Veerhuis R. Cerebrospinal fluid and plasma clusterin levels in Parkinson's disease. Parkinsonism Relat Disord 2013; 19:1079-83. [DOI: 10.1016/j.parkreldis.2013.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 05/25/2013] [Accepted: 07/16/2013] [Indexed: 10/26/2022]
|
47
|
A protocol for the management of canine cerebrospinal fluid for the proteomic assessment of putative biomarkers. Vet J 2013; 197:836-41. [DOI: 10.1016/j.tvjl.2013.05.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 05/24/2013] [Accepted: 05/27/2013] [Indexed: 11/24/2022]
|
48
|
Smeyne M, Smeyne RJ. Glutathione metabolism and Parkinson's disease. Free Radic Biol Med 2013; 62:13-25. [PMID: 23665395 PMCID: PMC3736736 DOI: 10.1016/j.freeradbiomed.2013.05.001] [Citation(s) in RCA: 321] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 05/01/2013] [Accepted: 05/01/2013] [Indexed: 12/14/2022]
Abstract
It has been established that oxidative stress, defined as the condition in which the sum of free radicals in a cell exceeds the antioxidant capacity of the cell, contributes to the pathogenesis of Parkinson disease. Glutathione is a ubiquitous thiol tripeptide that acts alone or in concert with enzymes within cells to reduce superoxide radicals, hydroxyl radicals, and peroxynitrites. In this review, we examine the synthesis, metabolism, and functional interactions of glutathione and discuss how these relate to the protection of dopaminergic neurons from oxidative damage and its therapeutic potential in Parkinson disease.
Collapse
Affiliation(s)
- Michelle Smeyne
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, 901-595-3066
| | - Richard Jay Smeyne
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, 901-595-2830
| |
Collapse
|
49
|
Lewitt PA, Li J, Lu M, Beach TG, Adler CH, Guo L. 3-hydroxykynurenine and other Parkinson's disease biomarkers discovered by metabolomic analysis. Mov Disord 2013; 28:1653-60. [PMID: 23873789 DOI: 10.1002/mds.25555] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 04/30/2013] [Accepted: 05/10/2013] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) biomarkers are needed to enhance therapeutics research and to understand PD pathogenesis. Methods that simultaneously measure hundreds of small molecular-weight compounds-metabolomic analysis-"fingerprint" disease-specific alterations in individual compounds or metabolic pathways. Beyond a nontargeted search for PD biomarkers, we hypothesized that PD cerebrospinal fluid would show increased formation of the excitotoxin 3-hydroxykynurenine and diminished concentration of the antioxidant glutathione. Cerebrospinal fluid was collected at <4 hours postmortem from 48 pathologically-verified PD subjects and 57 comparably-aged controls. Assays involved ultra-high-performance liquid and gas chromatography linked to mass spectrometry. We used univariate techniques to determine fold-changes in concentrations of biochemicals; false-discovery rates were calculated to exclude spurious findings. Data was modeled using a Support Vector Machine for analyzing compounds selected by Welch's t test. Classification accuracy was determined by cross-validation. Of 243 structurally-identified biochemicals,19 compounds differentiated PD from controls at a 20% false-discovery level. In PD, mean 3-hydroxykynurenine concentration was increased by one-third, and mean oxidized glutathione was decreased by 40% (for each, P < .01). Four of the 19 compounds differentiating PD from controls were N-acetylated amino acids, suggesting a generalized alteration in N-acetylation activity. The Support Vector Machine classification model distinguished between groups at 83% sensitivity and 91% specificity for the learning data, and at 65% and 79% from cross-validation. In this study, the first for metabolomic profiling of PD cerebrospinal fluid, we found several novel biomarkers and offer new directions for recognizing disease-specific biochemical indicators. The findings support involvement of excitotoxicity and oxidative stress in the pathogenesis of PD.
Collapse
Affiliation(s)
- Peter A Lewitt
- Department of Neurology, Henry Ford Hospital, West Bloomfield, Michigan, USA; School of Medicine, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Maarouf CL, Beach TG, Adler CH, Malek-Ahmadi M, Kokjohn TA, Dugger BN, Walker DG, Shill HA, Jacobson SA, Sabbagh MN, Roher AE. Quantitative appraisal of ventricular cerebrospinal fluid biomarkers in neuropathologically diagnosed Parkinson's disease cases lacking Alzheimer's disease pathology. Biomark Insights 2013; 8:19-28. [PMID: 23533154 PMCID: PMC3603385 DOI: 10.4137/bmi.s11422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Identifying biomarkers that distinguish Parkinson’s disease (PD) from normal control (NC) individuals has the potential to increase diagnostic sensitivity for the detection of early-stage PD. A previous proteomic study identified potential biomarkers in postmortem ventricular cerebrospinal fluid (V-CSF) from neuropathologically diagnosed PD subjects lacking Alzheimer’s disease (AD) neuropathology. In the present study, we assessed these biomarkers as well as p-tau181, Aβ42, and S100B by ELISA in PD (n = 43) and NC (n = 49) cases. The p-tau181/Aβ42 ratio and ApoA-1 showed statistically significant differences between groups. Multiple regression analysis demonstrated that p-tau181/Aβ42 had a significant odds ratio: OR = 1.42 (95% confidence interval [CI], 1.12–1.84), P = 0.006. Among the molecules investigated, intriguing correlations were observed that require further investigation. Our results suggest coexistent AD CSF biomarkers within the PD group notwithstanding that it was selected to minimize AD neuropathological lesions.
Collapse
Affiliation(s)
- Chera L Maarouf
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, AZ, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|