1
|
Chelucci E, Scarfò G, Piccarducci R, Rizza A, Fusi J, Epifani F, Carpi S, Polini B, Betti L, Costa B, Taliani S, Cela V, Artini P, Daniele S, Martini C, Franzoni F. Sex Differences in Blood Accumulation of Neurodegenerative-Related Proteins and Antioxidant Responses to Regular Physical Exercise. J Mol Neurosci 2024; 74:105. [PMID: 39496982 PMCID: PMC11535074 DOI: 10.1007/s12031-024-02278-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024]
Abstract
Physical activity has been demonstrated to improve cognitive function, thereby preventing/slowing neurodegenerative diseases (NDs). Biological responses to physical activity and vulnerabilities to NDs are emerging to be gender-related. Herein, known ND-associated markers (β-amyloid, tau, α-synuclein), main sex steroid hormones, antioxidant responses, and key gene transcription modulators were evaluated in the blood of physically active and sedentary women and men. In our hands, females presented higher basal erythrocytes β-amyloid and α-synuclein amounts than males. Regular physical activity was able to significantly reduce the erythrocyte content of β-amyloid in females and the tau levels in males, suggesting that these differences may be mediated by organizational actions of sex steroid hormones during development. Furthermore, despite a comparable plasma antioxidant capability (AOC) between males and females, in the latter group, physical activity significantly enhances AOC versus peroxynitrite radicals only. Finally, regular physical activity modulated the levels of transcription factor Nrf2 in erythrocytes, as well as the plasma concentration of the microRNA miR-195 and miR-153, suggesting the promotion of antioxidant/autophagic processes associated with ND-related proteins. Overall, these results could shed light on how cerebral adaptations to physical activity differ between males and females, especially with regard to blood accumulation of ND proteins and mechanisms of antioxidant responses to regular exercise.
Collapse
Affiliation(s)
| | - Giorgia Scarfò
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Antonio Rizza
- Interventional Cardiology Division, Gaetano Pasquinucci Heart Hospital, Fondazione Toscana Gabriele Monasterio, Massa, Italy
| | - Jonathan Fusi
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Epifani
- Department of Juridical and Economic Sciences, Pegaso Telematic University, Fanfani Diagnostics and Health, Florence, Italy
| | - Sara Carpi
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze-CNR and Scuola Normale Superiore, Pisa, Italy
| | | | - Laura Betti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Vito Cela
- Division of Gynecology and Obstetrics, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Paolo Artini
- Division of Gynecology and Obstetrics, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Pisa, Italy.
| | | | - Ferdinando Franzoni
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
2
|
Gaetani L, Paolini Paoletti F, Mechelli A, Bellomo G, Parnetti L. Research advancement in fluid biomarkers for Parkinson's disease. Expert Rev Mol Diagn 2024; 24:885-898. [PMID: 39262126 DOI: 10.1080/14737159.2024.2403073] [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: 06/22/2024] [Revised: 08/07/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Diagnostic criteria for Parkinson's disease (PD) rely on clinical, mainly motor, features, implying that pre-motor phase cannot be accurately identified. To achieve a reliable early diagnosis, similar to what has been done for Alzheimer's disease (AD), a shift from clinical to biological identification of PD is being pursued. This shift has taken great advantage from the research on cerebrospinal fluid (CSF) biomarkers as they mirror the ongoing molecular pathogenic mechanisms taking place in PD, thus intercepting the disease timely with respect to clinical manifestations. AREAS COVERED CSF α-synuclein seed amplification assay (αS-SAA) has emerged as the most promising biomarker of α-synucleinopathy. CSF biomarkers reflecting AD-pathology and axonal damage (neurofilament light chain) and a novel marker of dopaminergic dysfunction (DOPA decarboxylase) add valuable diagnostic and prognostic information in the neurochemical characterization of PD. EXPERT OPINION A biological classification system of PD, encompassing pathophysiological and staging biomarkers, might ensure both early identification and prognostic characterization of the patients. This approach could allow for the best setting for disease-modifying treatments which are currently under investigation.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Alessandro Mechelli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Yang Y, Nie X, Wang Y, Sun J, Gao X, Zhang J. Evolving insights into erythrocytes in synucleinopathies. Trends Neurosci 2024; 47:693-707. [PMID: 39043489 DOI: 10.1016/j.tins.2024.06.005] [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: 05/02/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/25/2024]
Abstract
Synucleinopathies, including Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), are characterized by neuronal loss accompanied by α-synuclein (α-syn) accumulation in the brain. While research conventionally focused on brain pathology, there is growing interest in peripheral alterations. Erythrocytes, which are rich in α-syn, have emerged as a compelling site for synucleinopathies-related alterations. Erythrocyte-derived extracellular vesicles (EVs), containing pathological α-syn species, can traverse the blood-brain barrier (BBB) under certain conditions and the gastrointestinal tract, where α-syn and gut microbiota interact extensively. This review explores the accumulating evidence of erythrocyte involvement in synucleinopathies, as well as their potential in disease pathogenesis and diagnosis. Given their unique properties, erythrocytes and erythrocyte-derived EVs may also serve as an ideal therapeutic platform for treating synucleinopathies and beyond.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoqian Nie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, China
| | - Yajie Wang
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Zhejiang, China
| | - Jie Sun
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Zhejiang, China
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Ma ZL, Wang ZL, Zhang FY, Liu HX, Mao LH, Yuan L. Biomarkers of Parkinson's Disease: From Basic Research to Clinical Practice. Aging Dis 2024; 15:1813-1830. [PMID: 37815899 PMCID: PMC11272192 DOI: 10.14336/ad.2023.1005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by dopaminergic neuron loss and the formation of Lewy bodies, which are enriched with aggregated α-synuclein (α-syn). PD currently has no cure, but therapeutic strategies are available to alleviate symptoms. Early diagnosis can greatly improve therapeutic interventions, but the clinical diagnosis of PD remains challenging and depends mainly on clinical features and imaging tests. Efficient and specific biomarkers are crucial for the diagnosis, monitoring, and evaluation of PD. Here, we reviewed the biomarkers of PD in different tissues and biofluids, along with the current clinical biochemical detection methods. We found that the sensitivity and specificity of single biomarkers are limited, and selecting appropriate indicators for combined detection can improve the diagnostic accuracy of PD.
Collapse
Affiliation(s)
| | | | - Fei-yue Zhang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Hong-xun Liu
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Li-hong Mao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Lin Yuan
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Lu Z, Yu X, Li P, Wang Y, Deng Y, Li X, Wang C, Yu S. Correlations of erythrocytic oligomer α-synuclein levels with age, sex and clinical variables in patients with Parkinson's disease. Front Aging Neurosci 2024; 16:1437622. [PMID: 39144258 PMCID: PMC11322579 DOI: 10.3389/fnagi.2024.1437622] [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: 05/24/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Oligomeric alpha-synuclein in red blood cells (RBC-o-α-Syn) has been shown to be increased in patients with Parkinson's disease (PD). However, factors that affect RBC-o-α-Syn levels remain to be elucidated. The aim of this study is to analyze the correlations between RBC-o-α-Syn levels and the age, sex and different clinical variables of patients with PD. Methods 167 patients with PD and 119 healthy controls (HC) were enrolled in this study. The patients with PD were diagnosed based on the MDS clinical diagnostic criteria for PD. All participants were evaluated for their clinical characteristics. Western blot analysis was used to examine the molecular sizes of RBC-o-α-Syn. A newly established chemiluminescent immunoassay was used to measure RBC-o-α-Syn levels. Results Higher RBC-o-α-Syn levels were detected in PD patients than in HC subjects. The receiver operating characteristic (ROC) curve indicated that a cut off value of 55.29 ng/mg discriminated well between PD patients and HC subjects, with a sensitivity of 67.66% (95% CI: 60.24-74.29%), a specificity of 88.24% (95% CI: 81.22-92.86%), and an area under the curve (AUC) of 0.857. The levels of RBC-o-α-Syn were higher in female than male patients (p = 0.033). For different subtypes, the levels of RBC-o-α-Syn were higher in the MIX subtype than the tremor-dominant (TD) PD. In addition, the levels of RBC-o-α-Syn were higher in patients with than without cognitive impairment (p = 0.016), and negatively correlated with Mini-Mental State Examination (MMSE) scores (r = -0.156, p = 0.044). Conclusion Our study demonstrates that RBC-o-α-Syn levels in patients with PD are higher than those in HC subjects and affected by the sex and the severity of cognitive impairment.
Collapse
Affiliation(s)
- Zhe Lu
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiaohan Yu
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Pengjie Li
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yiming Wang
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yeyun Deng
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Li
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chaodong Wang
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shun Yu
- Department of Neurobiology and National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Yadav S, Deepika, Moar K, Kumar A, Khola N, Pant A, Kakde GS, Maurya PK. Reconsidering red blood cells as the diagnostic potential for neurodegenerative disorders. Biol Cell 2024; 116:e2400019. [PMID: 38822416 DOI: 10.1111/boc.202400019] [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: 02/22/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Red blood cells (RBCs) are usually considered simple cells and transporters of gases to tissues. HYPOTHESIS However, recent research has suggested that RBCs may have diagnostic potential in major neurodegenerative disorders (NDDs). RESULTS This review summarizes the current knowledge on changes in RBC in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and other NDDs. It discusses the deposition of neuronal proteins like amyloid-β, tau, and α-synuclein, polyamines, changes in the proteins of RBCs like band-3, membrane transporter proteins, heat shock proteins, oxidative stress biomarkers, and altered metabolic pathways in RBCs during neurodegeneration. It also highlights the comparison of RBC diagnostic markers to other in-market diagnoses and discusses the challenges in utilizing RBCs as diagnostic tools, such as the need for standardized protocols and further validation studies. SIGNIFICANCE STATEMENT The evidence suggests that RBCs have diagnostic potential in neurodegenerative disorders, and this study can pave the foundation for further research which may lead to the development of novel diagnostic approaches and treatments.
Collapse
Affiliation(s)
- Somu Yadav
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Deepika
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Akshay Kumar
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Nikhila Khola
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Ganseh S Kakde
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| |
Collapse
|
7
|
Zhang J, Xie D, Jiao D, Zhou S, Liu S, Ju Z, Hu L, Qi L, Yao C, Zhao C. From inflammatory signaling to neuronal damage: Exploring NLR inflammasomes in ageing neurological disorders. Heliyon 2024; 10:e32688. [PMID: 38975145 PMCID: PMC11226848 DOI: 10.1016/j.heliyon.2024.e32688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
The persistence of neuronal degeneration and damage is a major obstacle in ageing medicine. Nucleotide-binding oligomerization domain (NOD)-like receptors detect environmental stressors and trigger the maturation and secretion of pro-inflammatory cytokines that can cause neuronal damage and accelerate cell death. NLR (NOD-like receptors) inflammasomes are protein complexes that contain NOD-like receptors. Studying the role of NLR inflammasomes in ageing-related neurological disorders can provide valuable insights into the mechanisms of neurodegeneration. This includes investigating their activation of inflammasomes, transcription, and capacity to promote or inhibit inflammatory signaling, as well as exploring strategies to regulate NLR inflammasomes levels. This review summarizes the use of NLR inflammasomes in guiding neuronal degeneration and injury during the ageing process, covering several neurological disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, stroke, and peripheral neuropathies. To improve the quality of life and slow the progression of neurological damage, NLR-based treatment strategies, including inhibitor-related therapies and physical therapy, are presented. Additionally, important connections between age-related neurological disorders and NLR inflammasomes are highlighted to guide future research and facilitate the development of new treatment options.
Collapse
Affiliation(s)
- Jingwen Zhang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong Xie
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Danli Jiao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuang Zhou
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shimin Liu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, 200030, China
| | - Ziyong Ju
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Hu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Qi
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chongjie Yao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chen Zhao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
8
|
Liu C, Su Y, Ma X, Wei Y, Qiao R. How close are we to a breakthrough? The hunt for blood biomarkers in Parkinson's disease diagnosis. Eur J Neurosci 2024; 59:2563-2576. [PMID: 38379501 DOI: 10.1111/ejn.16290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/22/2024]
Abstract
Parkinson's disease (PD), being the second largest neurodegenerative disease, poses challenges in early detection, resulting in a lack of timely treatment options to effectively manage the disease. By the time clinical diagnosis becomes possible, more than 60% of dopamine neurons in the substantia nigra (SN) of patients have already degenerated. Therefore, early diagnosis or identification of warning signs is crucial for the prompt and timely beginning of the treatment. However, conducting invasive or complex diagnostic procedures on asymptomatic patients can be challenging, making routine blood tests a more feasible approach in such cases. Numerous studies have been conducted over an extended period to search for effective diagnostic biomarkers in blood samples. However, thus far, no highly effective biomarkers have been confirmed. Besides classical proteins like α-synuclein (α-syn), phosphorylated α-syn and oligomeric α-syn, other molecules involved in disease progression should also be given equal attention. In this review, we will not only discuss proposed biomarkers that are currently under investigation but also delve into the mechanisms underlying the disease, focusing on processes such as α-syn misfolding, intercellular transmission and the crossing of the blood-brain barrier (BBB). Our aim is to provide an updated overview of molecules based on these processes that may potentially serve as blood biomarkers.
Collapse
Affiliation(s)
- Cheng Liu
- Peking University Third Hospital, Beijing, China
| | - Yang Su
- Peking University Third Hospital, Beijing, China
| | - Xiaolong Ma
- Peking University Third Hospital, Beijing, China
| | - Yao Wei
- Peking University Third Hospital, Beijing, China
| | - Rui Qiao
- Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Jeong S, Shim KH, Kim D, Bae H, Jeong DE, Kang MJ, An SSA. Assessment of acetylcholinesterase activity in CD9-positive exosomes from patients with Parkinson's disease. Front Aging Neurosci 2024; 16:1332455. [PMID: 38384937 PMCID: PMC10879351 DOI: 10.3389/fnagi.2024.1332455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Introduction Parkinson's disease (PD) is a neurodegenerative disorder characterized by dopaminergic dysfunction and associated with abnormalities in the cholinergic system. However, the relationship between PD and cholinergic dysfunction, particularly in exosomes, is not fully understood. Methods We enrolled 37 patients with PD and 44 healthy controls (HC) to investigate acetylcholinesterase (AChE) activity in CD9-positive and L1CAM-positive exosomes. Exosomes were isolated from plasma using antibody-coupled magnetic beads, and their sizes and concentrations were assessed using transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Subsequently, the AChE activity in these exosomes was analyzed in relation to various clinical parameters. Results A significant decrease in AChE activity was observed in CD9-positive exosomes derived from patients with PD, whereas no significant differences were found in L1CAM-positive exosomes. Further analysis with a larger sample size confirmed a substantial reduction in AChE activity in CD9-positive exosomes from the PD plasma, with moderate diagnostic accuracy. The decrease in AChE activity of CD9-positive exosomes did not show an association with cognitive impairment but displayed a trend toward correlation with PD progression. Discussion The reduction in AChE activity in CD9-positive exosomes suggests potential peripheral cholinergic dysfunction in PD, independent of the central cholinergic system. The observed alterations in AChE activity provide valuable insights into the association between cholinergic dysfunction and the pathogenesis of PD.
Collapse
Affiliation(s)
- Sumin Jeong
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
- Department of Neurology, Veterans Health Service Medical Center, Veterans Medical Research Institute, Seoul, Republic of Korea
| | - Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| | - Danyeong Kim
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
- Department of Neurology, Veterans Health Service Medical Center, Veterans Medical Research Institute, Seoul, Republic of Korea
| | - Heewon Bae
- Department of Neurology, Veterans Health Service Medical Center, Veterans Medical Research Institute, Seoul, Republic of Korea
| | - Da-Eun Jeong
- Department of Neurology, Veterans Health Service Medical Center, Veterans Medical Research Institute, Seoul, Republic of Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Health Service Medical Center, Veterans Medical Research Institute, Seoul, Republic of Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
10
|
Vijiaratnam N, Foltynie T. How should we be using biomarkers in trials of disease modification in Parkinson's disease? Brain 2023; 146:4845-4869. [PMID: 37536279 PMCID: PMC10690028 DOI: 10.1093/brain/awad265] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023] Open
Abstract
The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson's disease has formed the backbone for a proposed staging system for incorporation in Parkinson's disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson's disease patients into trials (as distinct from patients with non-Parkinson's disease parkinsonism or non-Parkinson's disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson's disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer's disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson's disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson's disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson's disease.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
11
|
Abdelmoaty MM, Lu E, Kadry R, Foster EG, Bhattarai S, Mosley RL, Gendelman HE. Clinical biomarkers for Lewy body diseases. Cell Biosci 2023; 13:209. [PMID: 37964309 PMCID: PMC10644566 DOI: 10.1186/s13578-023-01152-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders characterized by pathologic aggregates of neural and glial α-synuclein (α-syn) in the form of Lewy bodies (LBs), Lewy neurites, and cytoplasmic inclusions in both neurons and glia. Two major classes of synucleinopathies are LB disease and multiple system atrophy. LB diseases include Parkinson's disease (PD), PD with dementia, and dementia with LBs. All are increasing in prevalence. Effective diagnostics, disease-modifying therapies, and therapeutic monitoring are urgently needed. Diagnostics capable of differentiating LB diseases are based on signs and symptoms which might overlap. To date, no specific diagnostic test exists despite disease-specific pathologies. Diagnostics are aided by brain imaging and cerebrospinal fluid evaluations, but more accessible biomarkers remain in need. Mechanisms of α-syn evolution to pathologic oligomers and insoluble fibrils can provide one of a spectrum of biomarkers to link complex neural pathways to effective therapies. With these in mind, we review promising biomarkers linked to effective disease-modifying interventions.
Collapse
Affiliation(s)
- Mai M Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emma G Foster
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
12
|
Angius F, Mocci I, Ercoli T, Loy F, Fadda L, Palmas MF, Cannas G, Manzin A, Defazio G, Carta AR. Combined measure of salivary alpha-synuclein species as diagnostic biomarker for Parkinson's disease. J Neurol 2023; 270:5613-5621. [PMID: 37552278 PMCID: PMC10576686 DOI: 10.1007/s00415-023-11893-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/09/2023]
Abstract
Parkinson's disease (PD) diagnosis is still vulnerable to bias, and a definitive diagnosis often relies on post-mortem neuropathological diagnosis. In this regard, alpha-synuclein (αsyn)-specific in vivo biomarkers remain a critical unmet need, based on its relevance in the neuropathology. Specifically, content changes in αsyn species such as total (tot-αsyn), oligomeric (o-αsyn), and phosphorylated (p-αsyn) within the cerebrospinal fluid (CSF) and peripheral fluids (i.e., blood and saliva) have been proposed as PD biomarkers possibly reflecting the neuropathological outcome. Here, we measured the p-αsyn levels in the saliva from 15 PD patients along with tot-αsyn, o-αsyn and their ratios, and compared the results with those from 23 healthy subjects (HS), matched per age and sex. We also calculated the optimal cutoff values for different αsyn species to provide information about their capability to discriminate PD from HS. We found that p-αsyn was the most abundant alpha-synuclein species in the saliva. While p-αsyn concentration did not differ between PD and HS when adjusted for total salivary proteins, the ratio p-αsyn/tot-αsyn was largely lower in PD patients than in HS. Moreover, the concentration of o-αsyn was increased in the saliva of PD patients, and tot-αsyn did not differ between PD and HS. The ROC curves indicated that no single αsyn form or ratio could provide an accurate diagnosis of PD. On the other hand, the ratio of different items, namely p-αsyn/tot-αsyn and o-αsyn, yielded more satisfactory diagnostic accuracy, suggesting that the combined measure of different species in the saliva may show more promises as a diagnostic means for PD.
Collapse
Affiliation(s)
- Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ignazia Mocci
- CNR Institute of Translational Pharmacology, Unit of Cagliari, Cagliari, Italy
| | - Tommaso Ercoli
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Francesco Loy
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Laura Fadda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Giada Cannas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giovanni Defazio
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.
- Department of Translational Biomedicine and Neuroscience, Aldo Moro University of Bari, Bari, Italy.
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
13
|
Taneva SG, Todinova S, Andreeva T. Morphometric and Nanomechanical Screening of Peripheral Blood Cells with Atomic Force Microscopy for Label-Free Assessment of Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:14296. [PMID: 37762599 PMCID: PMC10531602 DOI: 10.3390/ijms241814296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegenerative disorders (NDDs) are complex, multifactorial disorders with significant social and economic impact in today's society. NDDs are predicted to become the second-most common cause of death in the next few decades due to an increase in life expectancy but also to a lack of early diagnosis and mainly symptomatic treatment. Despite recent advances in diagnostic and therapeutic methods, there are yet no reliable biomarkers identifying the complex pathways contributing to these pathologies. The development of new approaches for early diagnosis and new therapies, together with the identification of non-invasive and more cost-effective diagnostic biomarkers, is one of the main trends in NDD biomedical research. Here we summarize data on peripheral biomarkers, biofluids (cerebrospinal fluid and blood plasma), and peripheral blood cells (platelets (PLTs) and red blood cells (RBCs)), reported so far for the three most common NDDs-Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). PLTs and RBCs, beyond their primary physiological functions, are increasingly recognized as valuable sources of biomarkers for NDDs. Special attention is given to the morphological and nanomechanical signatures of PLTs and RBCs as biophysical markers for the three pathologies. Modifications of the surface nanostructure and morphometric and nanomechanical signatures of PLTs and RBCs from patients with AD, PD, and ALS have been revealed by atomic force microscopy (AFM). AFM is currently experiencing rapid and widespread adoption in biomedicine and clinical medicine, in particular for early diagnostics of various medical conditions. AFM is a unique instrument without an analog, allowing the generation of three-dimensional cell images with extremely high spatial resolution at near-atomic scale, which are complemented by insights into the mechanical properties of cells and subcellular structures. Data demonstrate that AFM can distinguish between the three pathologies and the normal, healthy state. The specific PLT and RBC signatures can serve as biomarkers in combination with the currently used diagnostic tools. We highlight the strong correlation of the morphological and nanomechanical signatures between RBCs and PLTs in PD, ALS, and AD.
Collapse
Affiliation(s)
- Stefka G. Taneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
| | - Tonya Andreeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. G. Bontchev” Str. 21, 1113 Sofia, Bulgaria; (S.T.); (T.A.)
- Faculty of Life Sciences, Reutlingen University, Alteburgstraße 150, D-72762 Reutlingen, Germany
| |
Collapse
|
14
|
Leitão AD, Spencer B, Sarsoza F, Ngolab J, Amalraj J, Masliah E, Wu C, Rissman RA. Hippocampal Reduction of α-Synuclein via RNA Interference Improves Neuropathology in Alzheimer's Disease Mice. J Alzheimers Dis 2023; 95:349-361. [PMID: 37522208 PMCID: PMC10578232 DOI: 10.3233/jad-230232] [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] [Accepted: 06/23/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) cases are often characterized by the pathological accumulation of α-synuclein (α-syn) in addition to amyloid-β (Aβ) and tau hallmarks. The role of α-syn has been extensively studied in synucleinopathy disorders, but less so in AD. Recent studies have shown that α-syn may also play a role in AD and its downregulation may be protective against the toxic effects of Aβ accumulation. OBJECTIVE We hypothesized that selectively knocking down α-syn via RNA interference improves the neuropathological and biochemical findings in AD mice. METHODS Here we used amyloid precursor protein transgenic (APP-Tg) mice to model AD and explore pathologic and behavioral phenotypes with knockdown of α-syn using RNA interference. We selectively reduced α-syn levels by stereotaxic bilateral injection of either LV-shRNA α-syn or LV-shRNA-luc (control) into the hippocampus of AD mice. RESULTS We found that downregulation of α-syn results in significant reduction in the number of Aβ plaques. In addition, mice treated with LV-shRNA α-syn had amelioration of abnormal microglial activation (Iba1) and astrocytosis (GFAP) phenotypes in AD mice. CONCLUSION Our data suggests a novel link between Aβ and α-syn pathology as well as a new therapeutic angle for targeting AD.
Collapse
Affiliation(s)
- André D.G. Leitão
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Floyd Sarsoza
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Jennifer Ngolab
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jessica Amalraj
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Robert A. Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Physiology and Neuroscience, Alzheimer’s Therapeutic Research Institute of the Keck School of Medicine of the University of Southern California, San Diego, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
15
|
Tönges L, Buhmann C, Klebe S, Klucken J, Kwon EH, Müller T, Pedrosa DJ, Schröter N, Riederer P, Lingor P. Blood-based biomarker in Parkinson's disease: potential for future applications in clinical research and practice. J Neural Transm (Vienna) 2022; 129:1201-1217. [PMID: 35428925 PMCID: PMC9463345 DOI: 10.1007/s00702-022-02498-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/27/2022] [Indexed: 12/12/2022]
Abstract
The clinical presentation of Parkinson's disease (PD) is both complex and heterogeneous, and its precise classification often requires an intensive work-up. The differential diagnosis, assessment of disease progression, evaluation of therapeutic responses, or identification of PD subtypes frequently remains uncertain from a clinical point of view. Various tissue- and fluid-based biomarkers are currently being investigated to improve the description of PD. From a clinician's perspective, signatures from blood that are relatively easy to obtain would have great potential for use in clinical practice if they fulfill the necessary requirements as PD biomarker. In this review article, we summarize the knowledge on blood-based PD biomarkers and present both a researcher's and a clinician's perspective on recent developments and potential future applications.
Collapse
Affiliation(s)
- Lars Tönges
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany.
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, 44801, Bochum, Nordrhein-Westfalen, Germany.
| | - Carsten Buhmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Stephan Klebe
- Department of Neurology, University Hospital Essen, 45147, Essen, Germany
| | - Jochen Klucken
- Department of Digital Medicine, University Luxembourg, LCSB, L-4367, Belval, Luxembourg
- Digital Medicine Research Group, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Centre Hospitalier de Luxembourg, Digital Medicine Research Clinic, L-1210, Luxembourg, Luxembourg
| | - Eun Hae Kwon
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany
| | - Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weissensee, 13088, Berlin, Germany
| | - David J Pedrosa
- Department of Neurology, Universitätsklinikum Gießen and Marburg, Marburg Site, 35043, Marburg, Germany
- Center of Mind, Brain and Behaviour (CMBB), Philipps-Universität Marburg, 35043, Marburg, Germany
| | - Nils Schröter
- Department of Neurology and Clinical Neuroscience, University of Freiburg, 79106, Freiburg, Germany
| | - Peter Riederer
- Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, 97080, Wuerzburg, Germany
- University of Southern Denmark Odense, 5000, Odense, Denmark
| | - Paul Lingor
- School of Medicine, Klinikum Rechts Der Isar, Department of Neurology, Technical University of Munich, 81675, München, Germany
| |
Collapse
|
16
|
Opportunities and challenges of alpha-synuclein as a potential biomarker for Parkinson's disease and other synucleinopathies. NPJ Parkinsons Dis 2022; 8:93. [PMID: 35869066 PMCID: PMC9307631 DOI: 10.1038/s41531-022-00357-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD), the second most common progressive neurodegenerative disease, develops and progresses for 10–15 years before the clinical diagnostic symptoms of the disease are manifested. Furthermore, several aspects of PD pathology overlap with other neurodegenerative diseases (NDDs) linked to alpha-synuclein (aSyn) aggregation, also called synucleinopathies. Therefore, there is an urgent need to discover and validate early diagnostic and prognostic markers that reflect disease pathophysiology, progression, severity, and potential differences in disease mechanisms between PD and other NDDs. The close association between aSyn and the development of pathology in synucleinopathies, along with the identification of aSyn species in biological fluids, has led to increasing interest in aSyn species as potential biomarkers for early diagnosis of PD and differentiate it from other synucleinopathies. In this review, we (1) provide an overview of the progress toward mapping the distribution of aSyn species in the brain, peripheral tissues, and biological fluids; (2) present comparative and critical analysis of previous studies that measured total aSyn as well as other species such as modified and aggregated forms of aSyn in different biological fluids; and (3) highlight conceptual and technical gaps and challenges that could hinder the development and validation of reliable aSyn biomarkers; and (4) outline a series of recommendations to address these challenges. Finally, we propose a combined biomarker approach based on integrating biochemical, aggregation and structure features of aSyn, in addition to other biomarkers of neurodegeneration. We believe that capturing the diversity of aSyn species is essential to develop robust assays and diagnostics for early detection, patient stratification, monitoring of disease progression, and differentiation between synucleinopathies. This could transform clinical trial design and implementation, accelerate the development of new therapies, and improve clinical decisions and treatment strategies.
Collapse
|
17
|
Cressatti M, Schipper HM. Dysregulation of a Heme Oxygenase-Synuclein Axis in Parkinson Disease. NEUROSCI 2022; 3:284-299. [PMID: 39483365 PMCID: PMC11523740 DOI: 10.3390/neurosci3020020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2024] Open
Abstract
α-Synuclein is a key driver of the pathogenesis of Parkinson disease (PD). Heme oxygenase-1 (HO-1), a stress protein that catalyzes the conversion of heme to biliverdin, carbon monoxide and free ferrous iron, is elevated in PD-affected neural tissues and promotes iron deposition and mitochondrial dysfunction in models of the disease, pathways also impacted by α-synuclein. Elevated expression of human HO-1 in astrocytes of GFAP.HMOX1 transgenic mice between 8.5 and 19 months of age elicits a parkinsonian phenotype characterized by nigrostriatal hypodopaminergia, locomotor incoordination and overproduction of neurotoxic native S129-phospho-α-synuclein. Two microRNAs (miRNA) known to regulate α-synuclein, miR-153 and miR-223, are significantly decreased in the basal ganglia of GFAP.HMOX1 mice. Serum concentrations of both miRNAs progressively decline in wild-type (WT) and GFAP.HMOX1 mice between 11 and 18 months of age. Moreover, circulating levels of miR-153 and miR-223 are significantly lower, and erythrocyte α-synuclein concentrations are increased, in GFAP.HMOX1 mice relative to WT values. MiR-153 and miR-223 are similarly decreased in the saliva of PD patients compared to healthy controls. Upregulation of glial HO-1 may promote parkinsonism by suppressing miR-153 and miR-223, which, in turn, enhance production of neurotoxic α-synuclein. The aim of the current review is to explore the link between HO-1, α-synuclein and PD, evaluating evidence derived from our laboratory and others. HO-1, miR-153 and miR-223 and α-synuclein may serve as potential biomarkers and targets for disease-modifying therapy in idiopathic PD.
Collapse
Affiliation(s)
- Marisa Cressatti
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| | - Hyman M Schipper
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| |
Collapse
|
18
|
Abstract
Amyloid-β (Aβ) peptides are involved in Alzheimer's disease (AD) development. The interactions of these peptides with copper and zinc ions also seem to be crucial for this pathology. Although Cu(II) and Zn(II) ions binding by Aβ peptides has been scrupulously investigated, surprisingly, this phenomenon has not been so thoroughly elucidated for N-truncated Aβ4-x-probably the most common version of this biomolecule. This negligence also applies to mixed Cu-Zn complexes. From the structural in silico analysis presented in this work, it appears that there are two possible mixed Cu-Zn(Aβ4-x) complexes with different stoichiometries and, consequently, distinct properties. The Cu-Zn(Aβ4-x) complex with 1:1:1 stoichiometry may have a neuroprotective superoxide dismutase-like activity. On the other hand, another mixed 2:1:2 Cu-Zn(Aβ4-x) complex is perhaps a seed for toxic oligomers. Hence, this work proposes a novel research direction for our better understanding of AD development.
Collapse
|
19
|
Ling L, Wang F, Yu D. Beyond neurodegenerative diseases: α-synuclein in erythropoiesis. Hematology 2022; 27:629-635. [PMID: 35621991 DOI: 10.1080/16078454.2022.2078041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
α-synuclein (α-syn) is a highly conserved and thermostable protein that is widely distributed in human brain. An intracellular aggregation of α-syn in dopaminergic neurons is the hallmark of a group of neurodegenerative diseases including Parkinson's disease. Interestingly, α-syn is also highly expressed in red blood cells and is considered as one of the most abundant proteins in red blood cells. Moreover, α-syn is thought to play a regulatory role during normal erythropoiesis. However, whether α-syn participates in the pathogenesis of erythroid diseases has not been reported. In this review, we discuss the protein structure of α-syn and the importance of α-syn in erythropoiesis.
Collapse
Affiliation(s)
- Ling Ling
- Institute of Translational Medicine, Yangzhou University, Medical College, Yangzhou, People's Republic of China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, People's Republic of China
| | - Fangfang Wang
- Institute of Translational Medicine, Yangzhou University, Medical College, Yangzhou, People's Republic of China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, People's Republic of China.,Department of Hematology, Yangzhou University, Clinical Medical College, Yangzhou, People's Republic of China
| | - Duonan Yu
- Institute of Translational Medicine, Yangzhou University, Medical College, Yangzhou, People's Republic of China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
20
|
Beatino MF, De Luca C, Campese N, Belli E, Piccarducci R, Giampietri L, Martini C, Perugi G, Siciliano G, Ceravolo R, Vergallo A, Hampel H, Baldacci F. α-synuclein as an emerging pathophysiological biomarker of Alzheimer's disease. Expert Rev Mol Diagn 2022; 22:411-425. [PMID: 35443850 DOI: 10.1080/14737159.2022.2068952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION α-syn aggregates represent the pathological hallmark of synucleinopathies as well as a frequent copathology (almost 1/3 of cases) in AD. Recent research indicates a potential role of α-syn species, measured in CSF with conventional analytical techniques, in the differential diagnosis between AD and synucleinopathies (such as DLB). Pioneering studies report the detection of α-syn in blood, however, conclusive investigations are controversial. Ultrasensitive seed amplification techniques, enabling the selective quantification of α-syn seeds, may represent an effective solution to identify the α-syn component in AD and facilitate a biomarker-guided stratification. AREAS COVERED We performed a PubMed-based review of the latest findings on α-syn-related biomarkers for AD, focusing on bodily fluids. A dissertation on the role of ultrasensitive seed amplification assays, detecting α-syn seeds from different biological samples, was conducted. EXPERT OPINION α-syn may contribute to progressive AD neurodegeneration through cross-seeding especially with tau protein. Ultrasensitive seed amplification techniques may support a biomarker-drug co-development pathway and may be a pathophysiological candidate biomarker for the evolving ATX(N) system to classify AD and the spectrum of primary NDDs. This would contribute to a precise approach to AD, aimed at implementing disease-modifying treatments.
Collapse
Affiliation(s)
| | - Ciro De Luca
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Giulio Perugi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| |
Collapse
|
21
|
Liu Z, Chan RB, Cai Z, Liu X, Wu Y, Yu Z, Feng T, Yang Y, Zhang J. α-Synuclein-containing erythrocytic extracellular vesicles: essential contributors to hyperactivation of monocytes in Parkinson's disease. J Neuroinflammation 2022; 19:53. [PMID: 35193594 PMCID: PMC8862590 DOI: 10.1186/s12974-022-02413-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/09/2022] [Indexed: 01/06/2023] Open
Abstract
Background Immune system dysfunction, including higher levels of peripheral monocytes and inflammatory cytokines, is an important feature of Parkinson’s disease (PD) pathogenesis, although the mechanism underlying the process remains to be investigated. In the central nervous system, it is well-known that α-synuclein (α-syn), a key protein involved in PD, activates microglia potently, and it is also reported that α-syn exists in the peripheral system, especially in erythrocytes or red blood cells (RBC) at exceedingly high concentration. The current study focused on the possibility that RBC-derived α-syn mediates the sensitization of peripheral monocytes in PD patients. Methods The hyperactivation of monocytes was assessed quantitatively by measuring mRNA levels of typical inflammatory cytokines (including IL-1β, IL-6 and TNF-α) and protein levels of secreted inflammatory cytokines (including pro-inflammatory cytokines: IL-1β, IL-6, TNF-α, IL-8, IFN-γ, IL-2, and IL-12p70 and anti-inflammatory cytokines: IL-4, IL-10, and IL-13). Western blot, nanoparticle tracking analysis and electron microscopy were used to characterize RBC-derived extracellular vesicles (RBC-EVs). Inhibitors of endocytosis and leucine-rich repeat kinase 2 (LRRK2), another key protein involved in PD, were used to investigate how these two factors mediated the process of monocyte sensitization by RBC-EVs. Results Increased inflammatory sensitization of monocytes was observed in PD patients and PD model mice. We found that α-syn-containing RBC-EVs isolated from PD model mice or free form oligomeric α-syn induced the inflammatory sensitization of THP-1 cells, and demonstrated that endocytosis was a requirement for this pathophysiological pathway. Furthermore, the hyperactivation of THP-1 cells induced by RBC-EVs was associated with increased LRRK2 production and kinase activity. The phenomenon of inflammatory sensitization of human monocytes and increased LRRK2 were also observed by the treatment of RBC-EVs isolated from PD patients. Conclusions Our data provided new insight into how hyperactivation of monocytes occurs in PD patients, and identified the central role played by α-syn-containing RBC-EVs in this process. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02413-1.
Collapse
Affiliation(s)
- Zongran Liu
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | | | - Zhijian Cai
- School of Basic Medicine, Zhejiang University, Hangzhou, 310002, Zhejiang, China
| | - Xiaodan Liu
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | - Yufeng Wu
- Department of Laboratory Medicine, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100050, China
| | - Tao Feng
- Department of Neurology, TianTan Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Yang
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China.
| | - Jing Zhang
- Department of Pathology, Zhejiang University School of Medicine and First Affiliated Hospital, Hangzhou, 310002, Zhejiang, China. .,National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
22
|
Cela V, Malacarne E, Obino MER, Marzi I, Papini F, Vergine F, Pisacreta E, Zappelli E, Pietrobono D, Scarfò G, Daniele S, Franzoni F, Martini C, Artini PG. Exploring Epithelial-Mesenchymal Transition Signals in Endometriosis Diagnosis and In Vitro Fertilization Outcomes. Biomedicines 2021; 9:biomedicines9111681. [PMID: 34829910 PMCID: PMC8615497 DOI: 10.3390/biomedicines9111681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 01/05/2023] Open
Abstract
Endometriosis (EMS) pathogenesis has been related to the release of inflammatory mediators in peritoneal fluid, creating an altered microenvironment that leads to low-grade oocyte/embryos and to the reduction of implantation rates. The Epithelial–Mesenchymal Transition (EMT), an inflammation-related process, can be a further contributing factor to EMS. This study aimed to investigate, among various cytokines and EMT markers (Cadherins, TGF-β, HIF-1α), diagnostic markers of EMS and prognostic factors of in vitro fertilization (IVF) outcomes. Herein, EMS patients manifested higher serum levels of the inflammatory molecules IL-6, IL-8, and IL-12 and a decrease in the concentrations of the anti-inflammatory IL-10. Moreover, biochemical markers associated with the EMT process were more elevated in serum and follicular fluid (FF) of EMS patients than in controls. At the end, the number of good-quality embryos was inversely related to serum IL-6 and EMT markers. Interestingly, serum IL-6 and FF IL-10 concentrations differentiated EMS patients from controls. Finally, serum IL-8 and E-Cadherin levels, as well as FF IL-10, predicted positive IVF outcome with great accuracy. Our data confirm the pivotal role of inflammatory mediators (i.e., IL-6 and IL-10) in EMS pathogenesis and suggest that EMT-related markers are elevated in EMS patients and can be predictive of IVF outcome.
Collapse
Affiliation(s)
- Vito Cela
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Elisa Malacarne
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Maria Elena Rosa Obino
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Ilaria Marzi
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Francesca Papini
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Francesca Vergine
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Elena Pisacreta
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
| | - Elisa Zappelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.Z.); (D.P.); (S.D.); (C.M.)
| | - Deborah Pietrobono
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.Z.); (D.P.); (S.D.); (C.M.)
| | - Giorgia Scarfò
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.S.); (F.F.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.Z.); (D.P.); (S.D.); (C.M.)
| | - Ferdinando Franzoni
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.S.); (F.F.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (E.Z.); (D.P.); (S.D.); (C.M.)
| | - Paolo Giovanni Artini
- Division of Gynecology and Obstetrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (V.C.); (E.M.); (M.E.R.O.); (I.M.); (F.P.); (F.V.); (E.P.)
- Correspondence: ; Tel.: +39-050-554-104
| |
Collapse
|
23
|
Li XY, Li W, Li X, Li XR, Sun L, Yang W, Cai Y, Chen Z, Wu J, Wang C, Yu S. Alterations of Erythrocytic Phosphorylated Alpha-Synuclein in Different Subtypes and Stages of Parkinson's Disease. Front Aging Neurosci 2021; 13:623977. [PMID: 34658833 PMCID: PMC8511781 DOI: 10.3389/fnagi.2021.623977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Serine 129-phosphorylated alpha-synuclein (pS-α-syn) is a major form of α-syn relevant to the pathogenesis of Parkinson's disease (PD), which has been recently detected in red blood cells (RBCs). However, alterations of RBC-derived pS-α-syn (pS-α-syn-RBC) in different subtypes and stages of PD remains to be investigated. In the present study, by using enzyme-linked immunosorbent assay (ELISA) to measure pS-α-syn-RBC, we demonstrated significantly higher levels of pS-α-syn-RBC in PD patients than in healthy controls. pS-α-syn-RBC separated the patients well from the controls, with a sensitivity of 93.39% (95% CI: 90.17-95.81%), a specificity of 93.11% (95% CI: 89.85-95.58%), and an area under the curve (AUC) of 0.96. Considering motor subtypes, the levels of pS-α-syn-RBC were significantly higher in late-onset than young-onset PD (p = 0.013) and in those with postural instability and gait difficulty than with tremor-dominant (TD) phenotype (p = 0.029). In addition, the levels of pS-α-syn-RBC were also different in non-motor subtypes, which were significantly lower in patients with cognitive impairment (p = 0.012) and olfactory loss (p = 0.004) than in those without such symptoms. Moreover, the levels of pS-α-syn-RBC in PD patients were positively correlated with disease duration and Hoehn & Yahr stages (H&Y) (p for trend =0.02 and <0.001) as well as UPDRS III (R 2 = 0.031, p = 0.0042) and MoCA scores (R 2 = 0.048, p = 0.0004). The results obtained suggest that pS-α-syn-RBC can be used as a potential biomarker for not only separating PD patients from healthy controls but also predicting the subtypes and stages of PD.
Collapse
Affiliation(s)
- Xu-Ying Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xin Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xu-Ran Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Linjuan Sun
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiwei Yang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhigang Chen
- Department of Neurology, Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Guangdong, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Amyloid particles facilitate surface-catalyzed cross-seeding by acting as promiscuous nanoparticles. Proc Natl Acad Sci U S A 2021; 118:2104148118. [PMID: 34462352 PMCID: PMC8433567 DOI: 10.1073/pnas.2104148118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Amyloid seeds are nanometer-sized protein particles that accelerate amyloid assembly as well as propagate and transmit the amyloid protein conformation associated with a wide range of protein misfolding diseases. However, seeded amyloid growth through templated elongation at fibril ends cannot explain the full range of molecular behaviors observed during cross-seeded formation of amyloid by heterologous seeds. Here, we demonstrate that amyloid seeds can accelerate amyloid formation via a surface catalysis mechanism without propagating the specific amyloid conformation associated with the seeds. This type of seeding mechanism is demonstrated through quantitative characterization of the cross-seeded assembly reactions involving two nonhomologous and unrelated proteins: the human Aβ42 peptide and the yeast prion-forming protein Sup35NM. Our results demonstrate experimental approaches to differentiate seeding by templated elongation from nontemplated amyloid seeding and rationalize the molecular mechanism of the cross-seeding phenomenon as a manifestation of the aberrant surface activities presented by amyloid seeds as nanoparticles.
Collapse
|
25
|
Ciobanu AM, Ionita I, Buleandra M, David IG, Popa DE, Ciucu AA, Budisteanu M. Current advances in metabolomic studies on non-motor psychiatric manifestations of Parkinson's disease (Review). Exp Ther Med 2021; 22:1010. [PMID: 34345292 PMCID: PMC8311266 DOI: 10.3892/etm.2021.10443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
Life expectancy has increased worldwide and, along with it, a greater prevalence of age-dependent disorders, chronic illnesses and comorbidities can be observed. In 2019, in both Europe and the Americas, dementias ranked 3rd among the top 10 causes of death. Parkinson's disease (PD) is the second most frequent type of neurodegenerative disease. In the last decades, globally, the number of people suffering from PD has more than doubled to over 6 million. Of all the neurological disorders, PD increased with the fastest rate. This troubling trend highlights the stringent need for accurate diagnostic biomarkers, especially in the early stages of the disease and to evaluate treatment response. To gain a broad and complex understanding of the recent advances in the '-omics' research fields, electronic databases such as PubMed, Google Academic, and Science Direct were searched for publications regarding metabolomic studies on PD to identify specific biomarkers for PD, and especially PD with associated psychiatric symptomatology. Discoveries in the fields of metagenomics, transcriptomics and proteomics, may lead to an improved comprehension of the metabolic pathways involved in disease etiology and progression and contribute to the discovery of novel therapeutic targets for effective treatment options.
Collapse
Affiliation(s)
- Adela Magdalena Ciobanu
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
- Department of Neurosciences, Discipline of Psychiatry, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ioana Ionita
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
| | - Mihaela Buleandra
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Iulia Gabriela David
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Dana Elena Popa
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Anton Alexandru Ciucu
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Magdalena Budisteanu
- Laboratory of Medical Genetics, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Medical Genetics, Faculty of Medicine, ‘Titu Maiorescu’ University, 031593 Bucharest, Romania
- Psychiatry Research Laboratory, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| |
Collapse
|
26
|
Ganguly U, Singh S, Pal S, Prasad S, Agrawal BK, Saini RV, Chakrabarti S. Alpha-Synuclein as a Biomarker of Parkinson's Disease: Good, but Not Good Enough. Front Aging Neurosci 2021; 13:702639. [PMID: 34305577 PMCID: PMC8298029 DOI: 10.3389/fnagi.2021.702639] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder of the elderly, presenting primarily with symptoms of motor impairment. The disease is diagnosed most commonly by clinical examination with a great degree of accuracy in specialized centers. However, in some cases, non-classical presentations occur when it may be difficult to distinguish the disease from other types of degenerative or non-degenerative movement disorders with overlapping symptoms. The diagnostic difficulty may also arise in patients at the early stage of PD. Thus, a biomarker could help clinicians circumvent such problems and help them monitor the improvement in disease pathology during anti-parkinsonian drug trials. This review first provides a brief overview of PD, emphasizing, in the process, the important role of α-synuclein in the pathogenesis of the disease. Various attempts made by the researchers to develop imaging, genetic, and various biochemical biomarkers for PD are then briefly reviewed to point out the absence of a definitive biomarker for this disorder. In view of the overwhelming importance of α-synuclein in the pathogenesis, a detailed analysis is then made of various studies to establish the biomarker potential of this protein in PD; these studies measured total α-synuclein, oligomeric, and post-translationally modified forms of α-synuclein in cerebrospinal fluid, blood (plasma, serum, erythrocytes, and circulating neuron-specific extracellular vesicles) and saliva in combination with certain other proteins. Multiple studies also examined the accumulation of α-synuclein in various forms in PD in the neural elements in the gut, submandibular glands, skin, and the retina. The measurements of the levels of certain forms of α-synuclein in some of these body fluids or their components or peripheral tissues hold a significant promise in establishing α-synuclein as a definitive biomarker for PD. However, many methodological issues related to detection and quantification of α-synuclein have to be resolved, and larger cross-sectional and follow-up studies with controls and patients of PD, parkinsonian disorders, and non-parkinsonian movement disorders are to be undertaken.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Sukhpal Singh
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Soumya Pal
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Suvarna Prasad
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Bimal K. Agrawal
- Department of General Medicine, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Reena V. Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| |
Collapse
|
27
|
Youssef P, Kim WS, Halliday GM, Lewis SJG, Dzamko N. Comparison of Different Platform Immunoassays for the Measurement of Plasma Alpha-Synuclein in Parkinson's Disease Patients. JOURNAL OF PARKINSONS DISEASE 2021; 11:1761-1772. [PMID: 34151860 PMCID: PMC8609717 DOI: 10.3233/jpd-212694] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: The identification of reliable biomarkers in Parkinson’s disease (PD) would provide much needed diagnostic accuracy, a means of monitoring progression, objectively measuring treatment response, and potentially allowing patient stratification within clinical trials. Whilst the assessment of total alpha-synuclein in biofluids has been identified as a promising biomarker, conflicting trends in these levels across patient plasma samples relative to controls has limited its use. Different commercially available assay platforms that have been used to measure alpha-synuclein may contribute to different study outcomes. Objective: To compare different platform immunoassays for the measurement of total alpha-synuclein using the same plasma samples from 49 PD patients and 47 controls. Methods: Total plasma alpha-synuclein concentrations were assessed using the BioLegend, MesoScale Discovery, and Quanterix platform in plasma samples from PD patients and matched controls. Results: A significant increase in total plasma alpha-synuclein was observed in PD patients using the Biolegend (10%), Mesoscale Discovery (13%) and Quanterix (39%) assays. The Mesoscale Discovery and Quanterix assays showed the strongest correlations (r = 0.78, p < 0.0001) with each other, whilst the Quanterix platform demonstrated the lowest variation and highest effect size. Inclusion of age, sex and hemoglobin levels as covariates in the analysis of total alpha-synuclein improved the ability of all three immunoassays to detect a significant difference between patients and controls. Conclusion: All three immunoassays were sensitive enough to detect group level differences between PD patients and controls, with the largest effect size observed with the Quanterix assay. These results may help inform assay choices in ongoing clinical trials.
Collapse
Affiliation(s)
- Priscilla Youssef
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Woojin S Kim
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Glenda M Halliday
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Simon J G Lewis
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Nicolas Dzamko
- School of Medical Sciences, Faculty of Medicine and Health and the Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
28
|
Cheslow L, Snook AE, Waldman SA. Emerging targets for the diagnosis of Parkinson's disease: examination of systemic biomarkers. Biomark Med 2021; 15:597-608. [PMID: 33988462 DOI: 10.2217/bmm-2020-0654] [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] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease (PD) is a highly prevalent and irreversible neurodegenerative disorder that is typically diagnosed in an advanced stage. Currently, there are no approved biomarkers that reliably identify PD patients before they have undergone extensive neuronal damage, eliminating the opportunity for future disease-modifying therapies to intervene in disease progression. This unmet need for diagnostic and therapeutic biomarkers has fueled PD research for decades, but these efforts have not yet yielded actionable results. Recently, studies exploring mechanisms underlying PD progression have offered insights into multisystemic contributions to pathology, challenging the classic perspective of PD as a disease isolated to the brain. This shift in understanding has opened the door to potential new biomarkers from multiple sites in the body. This review focuses on emerging candidates for PD biomarkers in the context of current diagnostic approaches and multiple organ systems that contribute to disease.
Collapse
Affiliation(s)
- Lara Cheslow
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E Snook
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A Waldman
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
29
|
Campese N, Palermo G, Del Gamba C, Beatino MF, Galgani A, Belli E, Del Prete E, Della Vecchia A, Vergallo A, Siciliano G, Ceravolo R, Hampel H, Baldacci F. Progress regarding the context-of-use of tau as biomarker of Alzheimer's disease and other neurodegenerative diseases. Expert Rev Proteomics 2021; 18:27-48. [PMID: 33545008 DOI: 10.1080/14789450.2021.1886929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Tau protein misfolding and accumulation in toxic species is a critical pathophysiological process of Alzheimer's disease (AD) and other neurodegenerative disorders (NDDs). Tau biomarkers, namely cerebrospinal fluid (CSF) total-tau (t-tau), 181-phosphorylated tau (p-tau), and tau-PET tracers, have been recently embedded in the diagnostic criteria for AD. Nevertheless, the role of tau as a diagnostic and prognostic biomarker for other NDDs remains controversial.Areas covered: We performed a systematical PubMed-based review of the most recent advances in tau-related biomarkers for NDDs. We focused on papers published from 2015 to 2020 assessing the diagnostic or prognostic value of each biomarker.Expert opinion: The assessment of tau biomarkers in alternative easily accessible matrices, through the development of ultrasensitive techniques, represents the most significant perspective for AD-biomarker research. In NDDs, novel tau isoforms (e.g. p-tau217) or proteolytic fragments (e.g. N-terminal fragments) may represent candidate diagnostic and prognostic biomarkers and may help monitoring disease progression. Protein misfolding amplification assays, allowing the identification of different tau strains (e.g. 3 R- vs. 4 R-tau) in CSF, may constitute a breakthrough for the in vivo stratification of NDDs. Tau-PET may help tracking the spatial-temporal evolution of tau pathophysiology in AD but its application outside the AD-spectrum deserves further studies.
Collapse
Affiliation(s)
- Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Del Gamba
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard De L'hôpital, Sorbonne University, Paris, France
| |
Collapse
|
30
|
Xu MM, Zhou MT, Li SW, Zhen XC, Yang S. Glycoproteins as diagnostic and prognostic biomarkers for neurodegenerative diseases: A glycoproteomic approach. J Neurosci Res 2021; 99:1308-1324. [PMID: 33634546 DOI: 10.1002/jnr.24805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDs) are incurable and can develop progressively debilitating disorders, including dementia and ataxias. Alzheimer's disease and Parkinson's disease are the most common NDs that mainly affect the elderly people. There is an urgent need to develop new diagnostic tools so that patients can be accurately stratified at an early stage. As a common post-translational modification, protein glycosylation plays a key role in physiological and pathological processes. The abnormal changes in glycosylation are associated with the altered biological pathways in NDs. The pathogenesis-related proteins, like amyloid-β and microtubule-associated protein tau, have altered glycosylation. Importantly, specific glycosylation changes in cerebrospinal fluid, blood and urine are valuable for revealing neurodegeneration in the early stages. This review describes the emerging biomarkers based on glycoproteomics in NDs, highlighting the potential applications of glycoprotein biomarkers in the early detection of diseases, monitoring of the disease progression, and measurement of the therapeutic responses. The mass spectrometry-based strategies for characterizing glycoprotein biomarkers are also introduced.
Collapse
Affiliation(s)
- Ming-Ming Xu
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | | | - Shu-Wei Li
- Nanjing Apollomics Biotech, Inc., Nanjing, China
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shuang Yang
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Daniele S, Baldacci F, Piccarducci R, Palermo G, Giampietri L, Manca ML, Pietrobono D, Frosini D, Nicoletti V, Tognoni G, Giorgi FS, Lo Gerfo A, Petrozzi L, Cavallini C, Franzoni F, Ceravolo R, Siciliano G, Trincavelli ML, Martini C, Bonuccelli U. α-Synuclein Heteromers in Red Blood Cells of Alzheimer's Disease and Lewy Body Dementia Patients. J Alzheimers Dis 2021; 80:885-893. [PMID: 33579836 DOI: 10.3233/jad-201038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Red blood cells (RBCs) contain the majority of α-synuclein (α-syn) in blood, representing an interesting model for studying the peripheral pathological alterations proved in neurodegeneration. OBJECTIVE The current study aimed to investigate the diagnostic value of total α-syn, amyloid-β (Aβ1-42), tau, and their heteroaggregates in RBCs of Lewy body dementia (LBD) and Alzheimer's disease (AD) patients compared to healthy controls (HC). METHODS By the use of enzyme-linked immunosorbent assays, RBCs concentrations of total α-syn, Aβ1-42, tau, and their heteroaggregates (α-syn/Aβ1-42 and α-syn/tau) were measured in 27 individuals with LBD (Parkinson's disease dementia, n = 17; dementia with Lewy bodies, n = 10), 51 individuals with AD (AD dementia, n = 37; prodromal AD, n = 14), and HC (n = 60). RESULTS The total α-syn and tau concentrations as well as α-syn/tau heterodimers were significantly lower in the LBD group and the AD group compared with HC, whereas α-syn/Aβ1-42 concentrations were significantly lower in the AD dementia group only. RBC α-syn/tau heterodimers had a higher diagnostic accuracy for differentiating patients with LBD versus HC (AUROC = 0.80). CONCLUSION RBC α-syn heteromers may be useful for differentiating between neurodegenerative dementias (LBD and AD) and HC. In particular, RBC α-syn/tau heterodimers have demonstrated good diagnostic accuracy for differentiating LBD from HC. However, they are not consistently different between LBD and AD. Our findings also suggest that α-syn, Aβ1-42, and tau interact in vivo to promote the aggregation and accumulation of each other.
Collapse
Affiliation(s)
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Giovanni Palermo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maria Laura Manca
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Mathematics, University of Pisa, Pisa, Italy
| | | | - Daniela Frosini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Valentina Nicoletti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Sean Giorgi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa Lo Gerfo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lucia Petrozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
32
|
Ma LY, Tian Y, Pan CR, Chen ZL, Ling Y, Ren K, Li JS, Feng T. Motor Progression in Early-Stage Parkinson's Disease: A Clinical Prediction Model and the Role of Cerebrospinal Fluid Biomarkers. Front Aging Neurosci 2021; 12:627199. [PMID: 33568988 PMCID: PMC7868416 DOI: 10.3389/fnagi.2020.627199] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
Background: The substantial heterogeneity of clinical symptoms and lack of reliable progression markers in Parkinson's disease (PD) present a major challenge in predicting accurate progression and prognoses. Increasing evidence indicates that each component of the neurovascular unit (NVU) and blood-brain barrier (BBB) disruption may take part in many neurodegenerative diseases. Since some portions of CSF are eliminated along the neurovascular unit and across the BBB, disturbing the pathways may result in changes of these substances. Methods: Four hundred seventy-four participants from the Parkinson's Progression Markers Initiative (PPMI) study (NCT01141023) were included in the study. Thirty-six initial features, including general information, brief clinical characteristics and the current year's classical scale scores, were used to build five regression models to predict PD motor progression represented by the coming year's Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Part III score after redundancy removal and recursive feature elimination (RFE)-based feature selection. Then, a threshold range was added to the predicted value for more convenient model application. Finally, we evaluated the CSF and blood biomarkers' influence on the disease progression model. Results: Eight hundred forty-nine cases were included in the study. The adjusted R2 values of three different categories of regression model, linear, Bayesian and ensemble, all reached 0.75. Models of the same category shared similar feature combinations. The common features selected among the categories were the MDS-UPDRS Part III score, Montreal Cognitive Assessment (MOCA) and Rapid Eye Movement Sleep Behavior Disorder Questionnaire (RBDSQ) score. It can be seen more intuitively that the model can achieve certain prediction effect through threshold range. Biomarkers had no significant impact on the progression model within the data in the study. Conclusions: By using machine learning and routinely gathered assessments from the current year, we developed multiple dynamic models to predict the following year's motor progression in the early stage of PD. These methods will allow clinicians to tailor medical management to the individual and identify at-risk patients for future clinical trials examining disease-modifying therapies.
Collapse
Affiliation(s)
- Ling-Yan Ma
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yu Tian
- Engineering Research Center of Electronic Medical Record (EMR) and Intelligent Expert System, College of Biomedical Engineering and Instrument Science, Zhejiang University, Ministry of Education, Hangzhou, China
| | - Chang-Rong Pan
- Engineering Research Center of Electronic Medical Record (EMR) and Intelligent Expert System, College of Biomedical Engineering and Instrument Science, Zhejiang University, Ministry of Education, Hangzhou, China
| | | | - Yun Ling
- Gyenno Science Co. Ltd., Shenzhen, China
| | - Kang Ren
- Gyenno Science Co. Ltd., Shenzhen, China
| | - Jing-Song Li
- Engineering Research Center of Electronic Medical Record (EMR) and Intelligent Expert System, College of Biomedical Engineering and Instrument Science, Zhejiang University, Ministry of Education, Hangzhou, China
| | - Tao Feng
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
33
|
Piccarducci R, Daniele S, Polini B, Carpi S, Chico L, Fusi J, Baldacci F, Siciliano G, Bonuccelli U, Nieri P, Martini C, Franzoni F. Apolipoprotein E Polymorphism and Oxidative Stress in Human Peripheral Blood Cells: Can Physical Activity Reactivate the Proteasome System through Epigenetic Mechanisms? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8869849. [PMID: 33488947 PMCID: PMC7796851 DOI: 10.1155/2021/8869849] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is characterized by proteasome activity impairment, oxidative stress, and epigenetic changes, resulting in β-amyloid (Aβ) production/degradation imbalance. Apolipoprotein E (ApoE) is implicated in Aβ clearance, and particularly, the ApoE ε4 isoform predisposes to AD development. Regular physical activity is known to reduce AD progression. However, the impact of ApoE polymorphism and physical exercise on Aβ production and proteasome system activity has never been investigated in human peripheral blood cells, particularly in erythrocytes, an emerging peripheral model used to study biochemical alteration. Therefore, the influence of ApoE polymorphism on the antioxidant defences, amyloid accumulation, and proteasome activity was here evaluated in human peripheral blood cells depending on physical activity, to assess putative peripheral biomarkers for AD and candidate targets that could be modulated by lifestyle. Healthy subjects were enrolled and classified based on the ApoE polymorphism (by the restriction fragment length polymorphism technique) and physical activity level (Borg scale) and grouped into ApoE ε4/non-ε4 carriers and active/non-active subjects. The plasma antioxidant capability (AOC), the erythrocyte Aβ production/accumulation, and the nuclear factor erythroid 2-related factor 2 (Nrf2) mediated proteasome functionality were evaluated in all groups by the chromatographic and immunoenzymatic assay, respectively. Moreover, epigenetic mechanisms were investigated considering the expression of histone deacetylase 6, employing a competitive ELISA, and the modulation of two key miRNAs (miR-153-3p and miR-195-5p), through the miRNeasy Serum/Plasma Mini Kit. ApoE ε4 subjects showed a reduction in plasma AOC and an increase in the Nrf2 blocker, miR-153-3p, contributing to an enhancement of the erythrocyte concentration of Aβ. Physical exercise increased plasma AOC and reduced the amount of Aβ and its precursor, involving a reduced miR-153-3p expression and a miR-195-5p enhancement. Our data highlight the impact of the ApoE genotype on the amyloidogenic pathway and the proteasome system, suggesting the positive impact of physical exercise, also through epigenetic mechanisms.
Collapse
Affiliation(s)
- Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Beatrice Polini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
- NEST, Istituto di Nanoscienze, Consiglio Nazionale delle Ricerche, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Lucia Chico
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| |
Collapse
|
34
|
Camporesi E, Nilsson J, Brinkmalm A, Becker B, Ashton NJ, Blennow K, Zetterberg H. Fluid Biomarkers for Synaptic Dysfunction and Loss. Biomark Insights 2020; 15:1177271920950319. [PMID: 32913390 PMCID: PMC7444114 DOI: 10.1177/1177271920950319] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Synapses are the site for brain communication where information is transmitted between neurons and stored for memory formation. Synaptic degeneration is a global and early pathogenic event in neurodegenerative disorders with reduced levels of pre- and postsynaptic proteins being recognized as a core feature of Alzheimer's disease (AD) pathophysiology. Together with AD, other neurodegenerative and neurodevelopmental disorders show altered synaptic homeostasis as an important pathogenic event, and due to that, they are commonly referred to as synaptopathies. The exact mechanisms of synapse dysfunction in the different diseases are not well understood and their study would help understanding the pathogenic role of synaptic degeneration, as well as differences and commonalities among them and highlight candidate synaptic biomarkers for specific disorders. The assessment of synaptic proteins in cerebrospinal fluid (CSF), which can reflect synaptic dysfunction in patients with cognitive disorders, is a keen area of interest. Substantial research efforts are now directed toward the investigation of CSF synaptic pathology to improve the diagnosis of neurodegenerative disorders at an early stage as well as to monitor clinical progression. In this review, we will first summarize the pathological events that lead to synapse loss and then discuss the available data on established (eg, neurogranin, SNAP-25, synaptotagmin-1, GAP-43, and α-syn) and emerging (eg, synaptic vesicle glycoprotein 2A and neuronal pentraxins) CSF biomarkers for synapse dysfunction, while highlighting possible utilities, disease specificity, and technical challenges for their detection.
Collapse
Affiliation(s)
- Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology & Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| |
Collapse
|
35
|
Graham C, Santiago-Mugica E, Abdel-All Z, Li M, McNally R, Kalaria RN, Mukaetova-Ladinska EB. Erythrocytes as Biomarkers for Dementia: Analysis of Protein Content and Alpha-Synuclein. J Alzheimers Dis 2020; 71:569-580. [PMID: 31424413 DOI: 10.3233/jad-190567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Discovering biomarkers for dementia is a pivotal step toward successful early diagnosis and treatment. Although plasma biomarkers have been explored, no consensus has been reached. Alpha-synuclein (AS), a 14 kDa synaptic protein associated with several neurodegenerative diseases, exists natively within erythrocytes (ERC). This protein is characteristic of Lewy body diseases, in which it aggregates into toxic Lewy bodies. As ERC are implicated in dementia, they are a potential target for future biomarkers. OBJECTIVE The aims of this study were to assess AS levels within ERC and whether AS can be used as a peripheral biomarker to differentiate between dementia and aged matched healthy control subjects. METHODS A total of 114 samples (60 aging controls, 36 Alzheimer's disease, 12 vascular dementia (VaD) and 6 dementia with Lewy bodies (DLB) subjects) were analyzed. We used Bradford assay to measure protein concentration, indirect ELISA to detect levels of AS, and immunoblotting to identify AS composition. Data were analyzed with nonparametric tests. RESULTS AS oligomers were present in dementia blood samples, whereas in controls, AS was largely monomeric. There was a significant increase in AS levels in DLB whole blood (p = 0.005; Kruskal-Wallis test), with a sensitivity and specificity of 100.0% and 93.9%. Protein concentrations in ERC isolated at pH 5.7 were significantly increased in dementia patients compared to controls (17.58 versus 40.33μg/ml; p≤0.005; Mann-Whitney test). In the VaD group, the protein concentration in the pH5.7 ERC fraction had sensitivity and specificity of 91.7% and 62.1%. CONCLUSIONS ERC protein concentration and AS levels have a potential for development of a novel diagnostic dementia blood test.
Collapse
Affiliation(s)
- Charlotte Graham
- Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | | | - Zeinab Abdel-All
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Mosi Li
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Richard McNally
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Rajesh N Kalaria
- Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Elizabeta B Mukaetova-Ladinska
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK.,The Evington Centre, Leicestershire Partnership NHS Trust, Leicester General Hospital, Leicester, UK
| |
Collapse
|
36
|
Wang L, Wang G, Duan Y, Wang F, Lin S, Zhang F, Li H, Li A, Li H. A Comparative Study of the Diagnostic Potential of Plasma and Erythrocytic α-Synuclein in Parkinson's Disease. NEURODEGENER DIS 2020; 19:204-210. [PMID: 32485710 DOI: 10.1159/000506480] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disease characterized by intracellular α-synuclein (α-Syn) deposition. Alternation of the α-Syn expression level in plasma or erythrocytes may be used as a potential PD biomarker. However, no studies have compared their prognostic value directly with the same cohort. METHODS The levels of α-Syn in plasma and erythrocytes, obtained from 45 PD patients and 45 control subjects, were measured with enzyme-linked immunosorbent assay. Then, correlation and receiver operating characteristic curve (ROC) analysis were performed to characterize the predictive power of erythrocytic and plasma α-Syn. RESULTS Our results showed that α-Syn expression levels in both plasma and erythrocytes were significantly higher in PD patients than in control subjects (823.14 ± 257.79 vs. 297.10 ± 192.82 pg/mL, p < 0.0001 in plasma; 3,104.14 ± 143.03 vs. 2,944.82 ± 200.41 pg/mL, p < 0.001 in erythrocytes, respectively). The results of the ROC analysis suggested that plasma α-Syn exhibited better predictive power than erythrocytic α-Syn with a sensitivity of 80.0%, specificity of 97.7%, and a positive predictive value of 77.8%. The expression level of plasma α-Syn correlated well with the age of patients, H-Y stage, MoCA scale, and UPDRS motor scale. On the contrary, there was no correlation between erythrocytic α-Syn level and clinical parameters in this study. CONCLUSION Our results suggest that plasma α-Syn could be a specific and sensitive potential diagnostic biomarker for PD.
Collapse
Affiliation(s)
- Luxuan Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| | - Guowei Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| | - Yangyang Duan
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| | - Feng Wang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| | - Shaoqing Lin
- Department of Neurology, Brain Center of Sunshine Union Hospital, Sunshine Union Hospital, Weifang, China
| | - Fengting Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| | - Hui Li
- Department of Computer Science, Jiangsu Ocean University, Lianyungang, China
| | - Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, North Carolina, USA,
| | - Haining Li
- Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, China
| |
Collapse
|
37
|
Pellegrini C, Daniele S, Antonioli L, Benvenuti L, D’Antongiovanni V, Piccarducci R, Pietrobono D, Citi V, Piragine E, Flori L, Ippolito C, Segnani C, Palazon-Riquelme P, Lopez-Castejon G, Martelli A, Colucci R, Bernardini N, Trincavelli ML, Calderone V, Martini C, Blandizzi C, Fornai M. Prodromal Intestinal Events in Alzheimer's Disease (AD): Colonic Dysmotility and Inflammation Are Associated with Enteric AD-Related Protein Deposition. Int J Mol Sci 2020; 21:ijms21103523. [PMID: 32429301 PMCID: PMC7278916 DOI: 10.3390/ijms21103523] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence suggests that intestinal dysfunctions may represent early events in Alzheimer’s disease and contribute to brain pathology. This study examined the relationship between onset of cognitive impairment and colonic dysfunctions in a spontaneous AD model before the full development of brain pathology. SAMP8 mice underwent Morris water maze and assessment of faecal output at four, six and eight months of age. In vitro colonic motility was examined. Faecal and colonic Aβ, tau proteins, α-synuclein and IL-1β were assessed by ELISA. Colonic citrate synthase activity was assessed by spectrophotometry. Colonic NLRP3, caspase-1 and ASC expression were evaluated by Western blotting. Colonic eosinophil density and claudin-1 expression were evaluated by immunohistochemistry. The effect of Aβ on NLRP3 signalling and mitochondrial function was tested in cultured cells. Cognitive impairment and decreased faecal output occurred in SAMP8 mice from six months. When compared with SAMR1, SAMP8 animals displayed: (1) impaired in vitro colonic contractions; (2) increased enteric AD-related proteins, IL-1β, active-caspase-1 expression and eosinophil density; and (3) decreased citrate synthase activity and claudin-1 expression. In THP-1 cells, Aβ promoted IL-1β release, which was abrogated upon incubation with caspase-1 inhibitor or in ASC-/- cells. Aβ decreased mitochondrial function in THP-1 cells. In SAMP8, enteric AD-related proteins deposition, inflammation and impaired colonic excitatory neurotransmission, occurring before the full brain pathology development, could contribute to bowel dysmotility and represent prodromal events in AD.
Collapse
Affiliation(s)
- Carolina Pellegrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.A.); (L.B.); (V.D.); (M.F.)
| | - Laura Benvenuti
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.A.); (L.B.); (V.D.); (M.F.)
| | - Vanessa D’Antongiovanni
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.A.); (L.B.); (V.D.); (M.F.)
| | - Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Deborah Pietrobono
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Lorenzo Flori
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Chiara Ippolito
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.I.); (C.S.); (N.B.)
| | - Cristina Segnani
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.I.); (C.S.); (N.B.)
| | - Pablo Palazon-Riquelme
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9PL, UK; (P.P.-R.); (G.L.-C.)
| | - Gloria Lopez-Castejon
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9PL, UK; (P.P.-R.); (G.L.-C.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy;
| | - Nunzia Bernardini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.I.); (C.S.); (N.B.)
- Interdepartmental Research Centre “Nutraceuticals and Food for Health”, University of Pisa, 56126 Pisa, Italy
| | - Maria Letizia Trincavelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (S.D.); (R.P.); (D.P.); (V.C.); (E.P.); (L.F.); (A.M.); (M.L.T.); (V.C.)
- Correspondence: (C.M.); (C.B.); Tel.: +39-050-221-2115 (C.M.); +39-050-221-8753 (C.B.)
| | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.A.); (L.B.); (V.D.); (M.F.)
- Correspondence: (C.M.); (C.B.); Tel.: +39-050-221-2115 (C.M.); +39-050-221-8753 (C.B.)
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (L.A.); (L.B.); (V.D.); (M.F.)
| |
Collapse
|
38
|
Ashton NJ, Hye A, Rajkumar AP, Leuzy A, Snowden S, Suárez-Calvet M, Karikari TK, Schöll M, La Joie R, Rabinovici GD, Höglund K, Ballard C, Hortobágyi T, Svenningsson P, Blennow K, Zetterberg H, Aarsland D. An update on blood-based biomarkers for non-Alzheimer neurodegenerative disorders. Nat Rev Neurol 2020; 16:265-284. [PMID: 32322100 DOI: 10.1038/s41582-020-0348-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 01/11/2023]
Abstract
Cerebrospinal fluid analyses and neuroimaging can identify the underlying pathophysiology at the earliest stage of some neurodegenerative disorders, but do not have the scalability needed for population screening. Therefore, a blood-based marker for such pathophysiology would have greater utility in a primary care setting and in eligibility screening for clinical trials. Rapid advances in ultra-sensitive assays have enabled the levels of pathological proteins to be measured in blood samples, but research has been predominantly focused on Alzheimer disease (AD). Nonetheless, proteins that were identified as potential blood-based biomarkers for AD, for example, amyloid-β, tau, phosphorylated tau and neurofilament light chain, are likely to be relevant to other neurodegenerative disorders that involve similar pathological processes and could also be useful for the differential diagnosis of clinical symptoms. This Review outlines the neuropathological, clinical, molecular imaging and cerebrospinal fluid features of the most common neurodegenerative disorders outside the AD continuum and gives an overview of the current status of blood-based biomarkers for these disorders.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Abdul Hye
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Anto P Rajkumar
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK.,Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Stuart Snowden
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Suárez-Calvet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Catalonia, Spain.,Department of Neurology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Memory Research Unit, Lund University, Malmö, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kina Höglund
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Disease Research, Neurogeriatrics Division, Karolinska Institutet, Novum, Huddinge, Stockholm, Sweden
| | | | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Per Svenningsson
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK. .,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK. .,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.
| |
Collapse
|
39
|
O'Hara DM, Kalia SK, Kalia LV. Methods for detecting toxic α-synuclein species as a biomarker for Parkinson's disease. Crit Rev Clin Lab Sci 2020; 57:291-307. [PMID: 32116096 DOI: 10.1080/10408363.2019.1711359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder and is characterized by the accumulation of α-synuclein (α-syn) into insoluble aggregates known as Lewy bodies and Lewy neurites in the brain. However, prior to the formation of these large aggregates, α-syn forms oligomers and small fibrils, which are believed to be the pathogenic species leading to the death of neurons in the substantia nigra in disease. The majority of aggregated α-syn is phosphorylated, and it is thought that this post-translational modification may be critical in disease pathogenesis. Thus, early detection of the toxic forms of α-syn may provide a window of opportunity for an intervention to halt or slow the progression of neurodegeneration in PD. Expression of α-syn is not restricted to the central nervous system and the protein can be found elsewhere, including bodily fluids and peripheral tissues. This review will examine current methods for detecting toxic forms of α-syn in accessible biospecimens and outline emerging techniques that may provide reliable identification of biomarkers for PD.
Collapse
Affiliation(s)
- Darren M O'Hara
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Suneil K Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Surgery, Division of Neurosurgery, University of Toronto, Toronto, Canada
| | - Lorraine V Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada.,Department of Medicine, Division of Neurology, Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| |
Collapse
|
40
|
Piccarducci R, Daniele S, Fusi J, Chico L, Baldacci F, Siciliano G, Bonuccelli U, Franzoni F, Martini C. Impact of ApoE Polymorphism and Physical Activity on Plasma Antioxidant Capability and Erythrocyte Membranes. Antioxidants (Basel) 2019; 8:E538. [PMID: 31717561 PMCID: PMC6912376 DOI: 10.3390/antiox8110538] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
The allele epsilon 4 (ε4) of apolipoprotein E (ApoE) is the strongest genetic risk factor for Alzheimer's disease (AD). ApoE protein plays a pivotal role in the synthesis and metabolism of amyloid beta (Aβ), the major component of the extracellular plaques that constitute AD pathological hallmarks. Regular exercise is an important preventive/therapeutic tool in aging and AD. Nevertheless, the impact of physical exercise on the well-being of erythrocytes, a good model of oxidative stress and neurodegenerative processes, remains to be investigated, particularly depending on ApoE polymorphism. Herein, we evaluate the oxidative status, Aβ levels, and the membrane's composition of erythrocytes in a cohort of human subjects. In our hands, the plasma antioxidant capability (AOC), erythrocytes membrane fluidity, and the amount of phosphatidylcholine (PC) were demonstrated to be significantly decreased in the ApoE ε4 genotype and non-active subjects. In contrast, erythrocyte Aβ content and lipid peroxidation increased in ε4 carriers. Regular physical exercise was associated with an increased plasma AOC and membrane fluidity, as well as to a reduced amount of erythrocytes Aβ. Altogether, these data highlight the influence of the ApoE genotype on erythrocytes' well-being and confirm the positive impact of regular physical exercise.
Collapse
Affiliation(s)
- Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Lucia Chico
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| |
Collapse
|
41
|
Petrillo S, Schirinzi T, Di Lazzaro G, D'Amico J, Colona VL, Bertini E, Pierantozzi M, Mari L, Mercuri NB, Piemonte F, Pisani A. Systemic Activation of Nrf2 Pathway in Parkinson's Disease. Mov Disord 2019; 35:180-184. [DOI: 10.1002/mds.27878] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
- Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases Children's Hospital and Research Institute Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico(I.R.C.C.S.) Rome Italy
| | - Tommaso Schirinzi
- Unit of Muscular and Neurodegenerative Diseases Children's Hospital and Research Institute Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico(I.R.C.C.S.) Rome Italy
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - Giulia Di Lazzaro
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - Jessica D'Amico
- Unit of Muscular and Neurodegenerative Diseases Children's Hospital and Research Institute Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico(I.R.C.C.S.) Rome Italy
| | - Vito L. Colona
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - Enrico Bertini
- Unit of Muscular and Neurodegenerative Diseases Children's Hospital and Research Institute Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico(I.R.C.C.S.) Rome Italy
| | | | - Luisa Mari
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - Nicola B. Mercuri
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Fondazione Santa Lucia I.R.C.C.S Rome Italy
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases Children's Hospital and Research Institute Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico(I.R.C.C.S.) Rome Italy
| | - Antonio Pisani
- Neurology, Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Fondazione Santa Lucia I.R.C.C.S Rome Italy
| |
Collapse
|
42
|
Cressatti M, Song W, Turk AZ, Garabed LR, Benchaya JA, Galindez C, Liberman A, Schipper HM. Glial HMOX1 expression promotes central and peripheral α-synuclein dysregulation and pathogenicity in parkinsonian mice. Glia 2019; 67:1730-1744. [PMID: 31180611 DOI: 10.1002/glia.23645] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 01/04/2023]
Abstract
α-Synuclein is a key player in the pathogenesis of Parkinson disease (PD). Expression of human heme oxygenase-1 (HO-1) in astrocytes of GFAP.HMOX1 transgenic (TG) mice between 8.5 and 19 months of age results in a parkinsonian phenotype characterized by neural oxidative stress, nigrostriatal hypodopaminergia associated with locomotor incoordination, and overproduction of α-synuclein. We identified two microRNAs (miR-), miR-153 and miR-223, that negatively regulate α-synuclein in the basal ganglia of male and female GFAP.HMOX1 mice. Serum concentrations of both miRNAs progressively declined in the wild-type (WT) and GFAP.HMOX1 mice between 11 and 19 months of age. Moreover, at each time point surveyed, circulating levels of miR-153 were significantly lower in the TG animals compared to WT controls, while α-synuclein protein concentrations were elevated in erythrocytes of the GFAP.HMOX1 mice at 19 months of age relative to WT values. Primary WT neurons co-cultured with GFAP.HMOX1 astrocytes exhibited enhanced protein oxidation, mitophagy and apoptosis, aberrant expression of genes regulating the dopaminergic phenotype, and an imbalance in gene expression profiles governing mitochondrial fission and fusion. Many, but not all, of these neuronal abnormalities were abrogated by small interfering RNA (siRNA) knockdown of α-synuclein, implicating α-synuclein as a potent, albeit partial, mediator of HO-1's neurodystrophic effects in these parkinsonian mice. Overexpression of HO-1 in stressed astroglia has previously been documented in the substantia nigra of idiopathic PD and may promote α-synuclein production and toxicity by downmodulating miR-153 and/or miR-223 both within the CNS and in peripheral tissues.
Collapse
Affiliation(s)
- Marisa Cressatti
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Wei Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Ariana Z Turk
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Laurianne R Garabed
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Joshua A Benchaya
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Carmela Galindez
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Adrienne Liberman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hyman M Schipper
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada.,Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
43
|
High Levels of β-Amyloid, Tau, and Phospho-Tau in Red Blood Cells as Biomarkers of Neuropathology in Senescence-Accelerated Mouse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5030475. [PMID: 31281579 PMCID: PMC6590616 DOI: 10.1155/2019/5030475] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/25/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's Disease (AD) is the most common Neurodegenerative Disease (ND), primarily characterised by neuroinflammation, neuronal plaques of β-amyloid (Aβ), and neurofibrillary tangles of hyperphosphorylated tau. α-Synuclein (α-syn) and its heteroaggregates with Aβ and tau have been recently included among the neuropathological elements of NDs. These pathological traits are not restricted to the brain, but they reach peripheral fluids as well. In this sense, Red Blood Cells (RBCs) are emerging as a good model to investigate the biochemical alterations of aging and NDs. Herein, the levels of homo- and heteroaggregates of ND-related proteins were analysed at different stages of disease progression. In particular, a validated animal model of AD, the SAMP8 (Senescence-Accelerated Mouse-Prone) and its control strain SAMR1 (Senescence-Accelerated Mouse-Resistant) were used in parallel experiments. The levels of the aforementioned proteins and of the inflammatory marker interleukin-1β (IL-1β) were examined in both brain and RBCs of SAMP8 and SAMR1 at 6 and 8 months. Brain Aβ, tau, and phospho-tau (p-tau) were higher in SAMP8 mice than in control mice and increased with AD progression. Similar accumulation kinetics were found in RBCs, even if slower. By contrast, α-syn and its heterocomplexes (α-syn-Aβ and α-syn-tau) displayed different accumulation kinetics between brain tissue and RBCs. Both brain and peripheral IL-1β levels were higher in SAMP8 mice, but increased sooner in RBCs, suggesting that inflammation might initiate at a peripheral level before affecting the brain. In conclusion, these results confirm RBCs as a valuable model for monitoring neurodegeneration, suggesting peripheral Aβ, tau, and p-tau as potential early biomarkers of AD.
Collapse
|
44
|
Tian C, Liu G, Gao L, Soltys D, Pan C, Stewart T, Shi M, Xie Z, Liu N, Feng T, Zhang J. Erythrocytic α-Synuclein as a potential biomarker for Parkinson's disease. Transl Neurodegener 2019; 8:15. [PMID: 31123587 PMCID: PMC6521422 DOI: 10.1186/s40035-019-0155-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
Background Erythrocytes are a major source of peripheral α-synuclein (α-Syn). The goal of the current investigation is to evaluate erythrocytic total, oligomeric/aggregated, and phosphorylated α-Syn species as biomarkers of Parkinson’s disease (PD). PD and healthy control blood samples were collected along with extensive clinical history to determine whether total, phosphorylated, or aggregated α-Syn derived from erythrocytes (the major source of blood α-Syn) are more promising and consistent biomarkers for PD than are free α-Syn species in serum or plasma. Methods Using newly developed electrochemiluminescence assays, concentrations of erythrocytic total, aggregated and phosphorylated at Ser129 (pS129) α-Syn, separated into membrane and cytosolic components, were measured in 225 PD patients and 133 healthy controls and analyzed with extensive clinical measures. Results The total and aggregated α-Syn levels were significantly higher in the membrane fraction of PD patients compared to healthy controls, but without alterations in the cytosolic component. The pS129 level was remarkably higher in PD subjects than in controls in the cytosolic fraction, and to a lesser extent, higher in the membrane fraction. Combining age, erythrocytic membrane aggregated α-Syn, and cytosolic pS129 levels, a model generated by using logistic regression analysis was able to discriminate patients with PD from neurologically normal controls, with a sensitivity and a specificity of 72 and 68%, respectively. Conclusions These results suggest that total, aggregated and phosphorylated α-Syn levels are altered in PD erythrocytes and peripheral erythrocytic α-Syn is a potential PD biomarker that needs further validation. Electronic supplementary material The online version of this article (10.1186/s40035-019-0155-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chen Tian
- 1Department of Pathology, Peking University School of Basic Medical Sciences, Peking University, Beijing, China.,6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Genliang Liu
- 3Center for Neurodegenerative Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,4China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Liyan Gao
- 8Department of neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.,3Center for Neurodegenerative Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - David Soltys
- 6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Catherine Pan
- 6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Tessandra Stewart
- 6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Min Shi
- 6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Zhiying Xie
- 6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA
| | - Na Liu
- Department of Neurology, Peking University Third Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Research and Transformation on Neurodegenerative Diseases Biomarkers, Beijing, China
| | - Tao Feng
- 3Center for Neurodegenerative Disease, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,4China National Clinical Research Center for Neurological Diseases, Beijing, China.,5Parkinson's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Jing Zhang
- 1Department of Pathology, Peking University School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Pathology, Peking University Third Hospital, Peking University, Beijing, China.,6Department of Pathology, University of Washington School of Medicine, Seattle, WA USA.,Beijing Key Laboratory of Research and Transformation on Neurodegenerative Diseases Biomarkers, Beijing, China
| |
Collapse
|
45
|
Parnetti L, Gaetani L, Eusebi P, Paciotti S, Hansson O, El-Agnaf O, Mollenhauer B, Blennow K, Calabresi P. CSF and blood biomarkers for Parkinson's disease. Lancet Neurol 2019; 18:573-586. [PMID: 30981640 DOI: 10.1016/s1474-4422(19)30024-9] [Citation(s) in RCA: 397] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/21/2018] [Accepted: 01/15/2019] [Indexed: 01/09/2023]
Abstract
In the management of Parkinson's disease, reliable diagnostic and prognostic biomarkers are urgently needed. The diagnosis of Parkinson's disease mostly relies on clinical symptoms, which hampers the detection of the earliest phases of the disease-the time at which treatment with forthcoming disease-modifying drugs could have the greatest therapeutic effect. Reliable prognostic markers could help in predicting the response to treatments. Evidence suggests potential diagnostic and prognostic value of CSF and blood biomarkers closely reflecting the pathophysiology of Parkinson's disease, such as α-synuclein species, lysosomal enzymes, markers of amyloid and tau pathology, and neurofilament light chain. A combination of multiple CSF biomarkers has emerged as an accurate diagnostic and prognostic model. With respect to early diagnosis, the measurement of CSF α-synuclein aggregates is providing encouraging preliminary results. Blood α-synuclein species and neurofilament light chain are also under investigation because they would provide a non-invasive tool, both for early and differential diagnosis of Parkinson's disease versus atypical parkinsonian disorders, and for disease monitoring. In view of adopting CSF and blood biomarkers for improving Parkinson's disease diagnostic and prognostic accuracy, further validation in large independent cohorts is needed.
Collapse
Affiliation(s)
- Lucilla Parnetti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine, University of Perugia, Perugia, Italy.
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Paolo Eusebi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Silvia Paciotti
- Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine, University of Perugia, Perugia, Italy; Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Education City, Doha, Qatar
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany; University Medical Center, Department of Neurology, Göttingen, Germany
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Paolo Calabresi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
46
|
Wang Q, Wang C, Ji B, Zhou J, Yang C, Chen J. Hapln2 in Neurological Diseases and Its Potential as Therapeutic Target. Front Aging Neurosci 2019; 11:60. [PMID: 30949044 PMCID: PMC6437066 DOI: 10.3389/fnagi.2019.00060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 03/01/2019] [Indexed: 01/18/2023] Open
Abstract
Hyaluronan and proteoglycan link protein 2 (Hapln2) is important for the binding of chondroitin sulfate proteoglycans to hyaluronan. Hapln2 deficiency leads to the abnormal expression of extracellular matrix (ECM) proteins and dysfunctional neuronal conductivity, demonstrating the vital role of Hapln2 in these processes. Studies have revealed that Hapln2 promotes the aggregation of α-synuclein, thereby contributing to neurodegeneration in Parkinson’s disease (PD), and it was recently suggested to be in intracellular neurofibrillary tangles (NFTs). Additionally, the expression levels of Hapln2 showed lower in the anterior temporal lobes of individuals with schizophrenia than those of healthy subjects. Together, these studies implicate the involvement of Hapln2 in the pathological processes of neurological diseases. A better understanding of the function of Hapln2 in the central nervous system (CNS) will provide new insights into the molecular mechanisms of these diseases and help to establish promising therapeutic strategies. Herein, we review the recent progress in defining the role of Hapln2 in brain physiology and pathology.
Collapse
Affiliation(s)
- Qinqin Wang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Chunmei Wang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Bingyuan Ji
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Jiawei Zhou
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chunqing Yang
- Neurobiology Key Laboratory, Jining Medical University, Jining, China
| | - Jing Chen
- Neurobiology Key Laboratory, Jining Medical University, Jining, China.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
47
|
Sun HL, Sun BL, Chen DW, Chen Y, Li WW, Xu MY, Shen YY, Xu ZQ, Wang YJ, Bu XL. Plasma α-synuclein levels are increased in patients with obstructive sleep apnea syndrome. Ann Clin Transl Neurol 2019; 6:788-794. [PMID: 31020003 PMCID: PMC6469340 DOI: 10.1002/acn3.756] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/22/2022] Open
Abstract
Objective Obstructive sleep apnea syndrome (OSAS) is characterized by nocturnal intermittent hypoxemia and can increase the risk of Parkinson's disease. This study aimed to investigate the association between plasma α‐synuclein levels and hypoxia in the patients with OSAS. Methods We recruited 42 OSAS patients and 46 controls with simple snoring matched for age and gender. OSAS was diagnosed on the basis of the clinical symptoms as well as the nighttime polysomnography. Plasma total α‐synuclein and phosphorylated α‐synuclein levels were measured by ELISA kits. Results The OSAS patients had significant higher levels of plasma total α‐synuclein and phosphorylated α‐synuclein levels. Both of the above indexes were positively correlated with the apnea–hypopnea index and the oxygen desaturation index, while they were negatively correlated with the mean and lowest oxyhemoglobin saturations. Interpretation This study suggests that chronic intermittent hypoxia can increase the α‐synuclein levels, which may contribute to the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Hao-Lun Sun
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Bin-Lu Sun
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Dong-Wan Chen
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Yang Chen
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Wei-Wei Li
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Man-Yu Xu
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Ying-Ying Shen
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Zhi-Qiang Xu
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
| |
Collapse
|
48
|
Potential Diagnostic Value of Red Blood Cells α-Synuclein Heteroaggregates in Alzheimer's Disease. Mol Neurobiol 2019; 56:6451-6459. [PMID: 30826968 DOI: 10.1007/s12035-019-1531-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/15/2019] [Indexed: 12/31/2022]
Abstract
A plethora of complex misfolded protein combinations have been found in Alzheimer disease (AD) brains besides the classical pathological hallmarks. Recently, α-synuclein (α-syn) and its heterocomplexes with amyloid-β (Aβ) and tau have been suggested to be involved in the pathophysiological processes of neurodegenerative diseases. These pathological features are not limited to the brain, but can be also found in peripheral fluids. In this respect, red blood cells (RBCs) have been suggested as a good model to investigate the biochemical alterations of neurodegeneration. Our aim is to find whether RBC concentrations of α-syn and its heterocomplexes (i.e., α-syn/Aβ and α-syn/tau) were different in AD patients compared with healthy controls (HC). The levels of homo- and heteroaggregates of α-syn, Aβ and tau, were analyzed in a cohort of AD patients at early stage either with dementia or prodromal symptoms (N = 39) and age-matched healthy controls (N = 39). All AD patients received a biomarker-based diagnosis (low cerebrospinal fluid levels of Aβ peptide combined with high cerebrospinal fluid concentrations of total tau and/or phospho-tau proteins; alternatively, a positivity to cerebral amyloid-PET scan). Our results showed lower concentrations of α-syn and its heterocomplexes (i.e., α-syn/Aβ and α-syn/tau) in RBCs of AD patients with respect to HC. RBC α-syn/Aβ as well as RBC α-syn/tau heterodimers discriminated AD participants from HC with fair accuracy, whereas RBC α-syn concentrations differentiated poorly the two groups. Although additional investigations are required, these data suggest α-syn heteroaggregates in RBCs as potential tool in the diagnostic work-up of early AD diagnosis.
Collapse
|
49
|
Xie F, Gao X, Yang W, Chang Z, Yang X, Wei X, Huang Z, Xie H, Yue Z, Zhou F, Wang Q. Advances in the Research of Risk Factors and Prodromal Biomarkers of Parkinson's Disease. ACS Chem Neurosci 2019; 10:973-990. [PMID: 30590011 DOI: 10.1021/acschemneuro.8b00520] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. With the advent of an aging population and improving life expectancy worldwide, the number of PD patients is expected to increase, which may lead to an urgent need for effective preventive and diagnostic strategies for PD. Although there is increasing research regarding the pathogenesis of PD, there is limited knowledge regarding the prevention of PD. Moreover, the diagnosis of PD depends on clinical criteria, which require the occurrence of bradykinesia and at least one symptom of rest tremor or rigidity. However, converging evidence from clinical, genetic, neuropathological, and imaging studies suggests the initiation of PD-specific pathology prior to the initial presentation of these classical motor clinical features by years or decades. This latent stage of neurodegeneration in PD is a particularly important stage for effective neuroprotective therapies, which might retard the progression or prevent the onset of PD. Therefore, the exploration of risk factors and premotor biomarkers is not only crucial to the early diagnosis of PD but is also helpful in the development of effective neuroprotection and health care strategies for appropriate populations at risk for PD. In this review, we searched and summarized ∼249 researches and 31 reviews focusing on the risk factors and prodromal biomarkers of PD and published in MEDLINE.
Collapse
Affiliation(s)
- Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaohua Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Huifang Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Hess Research Center Ninth Floor, New York, New York 10029, United States
| | - Fengli Zhou
- Department of Respiratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Gongye Road 253, Guangzhou, Guangdong 510280, P. R. China
| |
Collapse
|
50
|
Kim A, Nigmatullina R, Zalyalova Z, Soshnikova N, Krasnov A, Vorobyeva N, Georgieva S, Kudrin V, Narkevich V, Ugrumov M. Upgraded Methodology for the Development of Early Diagnosis of Parkinson's Disease Based on Searching Blood Markers in Patients and Experimental Models. Mol Neurobiol 2018; 56:3437-3450. [PMID: 30128652 DOI: 10.1007/s12035-018-1315-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 08/10/2018] [Indexed: 01/08/2023]
Abstract
Numerous attempts to develop an early diagnosis of Parkinson's disease (PD) by searching biomarkers in biological fluids were unsuccessful. The drawback of this methodology is searching markers in patients at the clinical stage without guarantee that they are also characteristic of either preclinical stage or prodromal stage (preclinical-prodromal stage). We attempted to upgrade this methodology by selecting only markers that are found both in patients and in PD animal models. HPLC and RT-PCR were used to estimate the concentration of amino acids, catecholamines/metabolites in plasma and gene expression in lymphocytes in 36 untreated early-stage PD patients and 52 controls, and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice at modeling the clinical ("symptomatic") stage and preclinical-prodromal ("presymptomatic") stage of PD. It was shown that among 13 blood markers found in patients, 7 markers are characteristic of parkinsonian symptomatic mice and 3 markers of both symptomatic and presymptomatic mice. According to our suggestion, the detection of the same marker in patients and symptomatic animals indicates adequate reproduction of pathogenesis along the corresponding metabolic pathway, whereas the detection of the same marker in presymptomatic animals indicates its specificity for preclinical-prodromal stage. This means that the minority of markers found in patients-decreased concentration of L-3,4-dihydroxyphenylalanine (L-DOPA) and dihydroxyphenylacetic acid (DOPAC) and increased dopamine D3 receptor gene expression-are specific for preclinical-prodromal stage and are suitable for early diagnosis of PD. Thus, we upgraded a current methodology for development of early diagnosis of PD by searching blood markers not only in patients but also in parkinsonian animals.
Collapse
Affiliation(s)
- Alexander Kim
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Razina Nigmatullina
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia
| | - Zuleikha Zalyalova
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia
- Kazan Hospital for War Veterans, Ministry of Health of the Republic of Tatarstan, Kazan, Russia
| | | | - Alexey Krasnov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Sofia Georgieva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | - Michael Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
- National Research University Higher School of Economics, Moscow, Russia.
| |
Collapse
|