1
|
Huoshen W, Zhu H, Xiong J, Chen X, Mou Y, Hou S, Yang B, Yi S, He Y, Huang H, Sun C, Li C. Identification of Potential Biomarkers and Therapeutic Targets for Periodontitis. Int Dent J 2024:S0020-6539(24)01554-5. [PMID: 39532570 DOI: 10.1016/j.identj.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Periodontitis is a chronic and multifactorial inflammatory disease. However, existing medications often lack sufficient therapeutic effects. The aim is to identify potential biomarkers and efficient therapeutic targets using Mendelian randomisation (MR) and single-cell analysis. METHODS MR analysis was conducted based on the cis-expression quantitative trait loci (cis-eQTLs) extracted from the eQTLGen Consortium and genome-wide association study (GWAS) data of periodontitis sourced from the Gene Lifestyle Interactions in Dental Endpoints (GLIDE) consortium (17,353 cases, 28,210 controls). Subsequently, colocalisation analysis was employed to detect whether genes and periodontitis shared the same casual variant. Finally, enrichment analysis, protein-protein interaction (PPI) networks, drug prediction, phenome-wide association study (PheWAS), molecular docking, and single-cell analysis were conducted to validate the significance of target genes. RESULTS Fourteen drug targets were significant related with periodontitis in MR analysis. Following the colocalisation and summary-data-based MR (SMR) analysis, 3 targets (S100A12, S100A9, and S100A8) were classified into tier 1 with strong evidence, 6 therapeutic targets (ADAM12, ADHFE1, BLK, HEBP1, SERPINE2, and TEK) were classified into tier 2 with moderate evidence, and 5 therapeutic targets (LY86, MMEL1, S100B, SPP1, and TRIB3) were classified into tier 3 with convincing evidence. PheWAS analysis showed that only TEK and SPP1 in tier 2 may induce side effects, including cardiometabolic and oncological issues. Molecular docking demonstrated strong binding between drugs and their respective protein targets. In the single-cell analysis, 5 target genes (HEBP1, LY86, S100A8, S100A9, and S100A12) exhibited enrichment in monocytes, while BLK and LY86 were primarily enriched in B cells. CONCLUSION The study identified 14 potential therapeutic targets for periodontitis. Among these, 3 therapeutic targets (S100A12, S100A9, and S100A8) demonstrated robust and well-supported results. Drugs designed to target these genes have a higher possibility of success in clinical trials, which are hopeful for prioritising periodontitis drug development.
Collapse
Affiliation(s)
- Wuda Huoshen
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China; Liangshan Minority Middle School, Liangshan, Sichuan, China
| | - Hanfang Zhu
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Junkai Xiong
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinyu Chen
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Yunjie Mou
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Shuhan Hou
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Yang
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Sha Yi
- Liangshan Minority Middle School, Liangshan, Sichuan, China
| | - Yahan He
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Liangshan Minority Middle School, Liangshan, Sichuan, China
| | - Haonan Huang
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Sun
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Chunhui Li
- Department of Periodontics and Oral Mucosal Diseases, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China; Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Gasparotto J, Somensi N, Girardi CS, Bittencourt RR, de Oliveira LM, Hoefel LP, Scheibel IM, Peixoto DO, Moreira JCF, Outeiro TF, Gelain DP. Is it all the RAGE? Defining the role of the receptor for advanced glycation end products in Parkinson's disease. J Neurochem 2024; 168:1608-1624. [PMID: 37381043 DOI: 10.1111/jnc.15890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a transmembrane receptor that belongs to the immunoglobulin superfamily and is extensively associated with chronic inflammation in non-transmissible diseases. As chronic inflammation is consistently present in neurodegenerative diseases, it was largely assumed that RAGE could act as a critical modulator of neuroinflammation in Parkinson's disease (PD), similar to what was reported for Alzheimer's disease (AD), where RAGE is postulated to mediate pro-inflammatory signaling in microglia by binding to amyloid-β peptide. However, accumulating evidence from studies of RAGE in PD models suggests a less obvious scenario. Here, we review physiological aspects of RAGE and address the current questions about the potential involvement of this receptor in the cellular events that may be critical for the development and progression of PD, exploring possible mechanisms beyond the classical view of the microglial activation/neuroinflammation/neurodegeneration axis that is widely assumed to be the general mechanism of RAGE action in the adult brain.
Collapse
Affiliation(s)
- Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brazil
| | - Nauana Somensi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carolina Saibro Girardi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Reykla Ramon Bittencourt
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Martinewski de Oliveira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Piloneto Hoefel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ingrid Matsubara Scheibel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Oppermann Peixoto
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Claudio Fonseca Moreira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Daniel Pens Gelain
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
3
|
Zhao N, Wang J, Huang S, Zhang J, Bao J, Ni H, Gao X, Zhang C. The landscape of programmed cell death-related lncRNAs in Alzheimer's disease and Parkinson's disease. Apoptosis 2024:10.1007/s10495-024-01984-z. [PMID: 38853201 DOI: 10.1007/s10495-024-01984-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/11/2024]
Abstract
This study delivers a thorough analysis of long non-coding RNAs (lncRNAs) in regulating programmed cell death (PCD), vital for neurodegenerative diseases like Alzheimer's disease (AD) and Parkinson's disease (PD). We propose a new framework PCDLnc, and identified 20 significant lncRNAs, including HEIH, SNHG15, and SNHG5, associated with PCD gene sets, which were known for roles in proliferation and apoptosis in neurodegenerative diseases. By using GREAT software, we identified regulatory functions of top lncRNAs in different neurodegenerative diseases. Moreover, lncRNAs cis-regulated mRNAs linked to neurodegeneration, including JAK2, AKT1, EGFR, CDC42, SNCA, and ADIPOQ, highlighting their therapeutic potential in neurodegenerative diseases. A further exploration into the differential expression of mRNA identified by PCDLnc revealed a role in apoptosis, ferroptosis and autophagy. Additionally, protein-protein interaction (PPI) network analysis exposed abnormal interactions among key genes, despite their consistent expression levels between disease and normal samples. The randomforest model effectively distinguished between disease samples, indicating a high level of accuracy. Shared gene subsets in AD and PD might serve as potential biomarkers, along with disease-specific gene sets. Besides, we also found the strong relationship between AD and immune infiltration. This research highlights the role of lncRNAs and their associated genes in PCD in neurodegenerative diseases, offering potential therapeutic targets and diagnostic markers for future study and clinical application.
Collapse
Affiliation(s)
- Ning Zhao
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Junyi Wang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Shan Huang
- The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingyu Zhang
- The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jin Bao
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Haisen Ni
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Xinhang Gao
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Chunlong Zhang
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, Heilongjiang, China.
| |
Collapse
|
4
|
Chang NP, DaPrano EM, Lindman M, Estevez I, Chou TW, Evans WR, Nissenbaum M, McCourt M, Alzate D, Atkins C, Kusnecov AW, Huda R, Daniels BP. Neuronal DAMPs exacerbate neurodegeneration via astrocytic RIPK3 signaling. JCI Insight 2024; 9:e177002. [PMID: 38713518 PMCID: PMC11382884 DOI: 10.1172/jci.insight.177002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
Astrocyte activation is a common feature of neurodegenerative diseases. However, the ways in which dying neurons influence the activity of astrocytes is poorly understood. Receptor interacting protein kinase-3 (RIPK3) signaling has recently been described as a key regulator of neuroinflammation, but whether this kinase mediates astrocytic responsiveness to neuronal death has not yet been studied. Here, we used the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine model of Parkinson's disease to show that activation of astrocytic RIPK3 drives dopaminergic cell death and axon damage. Transcriptomic profiling revealed that astrocytic RIPK3 promoted gene expression associated with neuroinflammation and movement disorders, and this coincided with significant engagement of damage-associated molecular pattern signaling. In mechanistic experiments, we showed that factors released from dying neurons signaled through receptor for advanced glycation endproducts to induce astrocytic RIPK3 signaling, which conferred inflammatory and neurotoxic functional activity. These findings highlight a mechanism of neuron-glia crosstalk in which neuronal death perpetuates further neurodegeneration by engaging inflammatory astrocyte activation via RIPK3.
Collapse
Affiliation(s)
| | | | | | | | | | - Wesley R Evans
- Department of Cell Biology and Neuroscience
- W. M. Keck Center for Collaborative Neuroscience, and
| | | | | | | | | | | | - Rafiq Huda
- Department of Cell Biology and Neuroscience
- W. M. Keck Center for Collaborative Neuroscience, and
| | | |
Collapse
|
5
|
Berntson L, Elfving A, Samuelsson AG, Öman A, Mobarrez F. Blood brain barrier permeability and astrocyte-derived extracellular vesicles in children with juvenile idiopathic arthritis: a cross-sectional study. Pediatr Rheumatol Online J 2024; 22:47. [PMID: 38671467 PMCID: PMC11046815 DOI: 10.1186/s12969-024-00984-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is the most prevalent rheumatic disease in children, and the inflammatory process is widely studied, primarily characterized by its impact on joint health. Emerging evidence suggests that JIA may also affect the central nervous system (CNS). This study investigates the potential CNS involvement in JIA by analyzing the presence of astrocyte-derived extracellular vesicles (EVs) and the S100B protein in plasma, both of which are indicative of astrocyte activity and blood-brain barrier (BBB) integrity. METHODS EDTA plasma from 90 children diagnosed with JIA and 10 healthy controls, matched by age and gender, was analyzed for extracellular vesicles by flow cytometric measurement. Astrocyte-derived EVs were identified using flow cytometry with markers for aquaporin 4 (AQP-4) and glial fibrillary acidic protein (GFAP). Levels of the S100B protein were measured using a commercial ELISA. Disease activity was assessed using the Juvenile Arthritis Disease Activity Score (JADAS27, 0-57), and pain levels were measured using a visual analogue scale (VAS, 0-10 cm). RESULTS Our analyses revealed a significantly higher concentration of astrocyte-derived EVs in the plasma of children with JIA compared with healthy controls. Furthermore, children with JADAS27 scores of 1 or higher exhibited notably higher levels of these EVs. The S100B protein was detectable exclusively in the JIA group. CONCLUSION The elevated levels of astrocyte-derived EVs and the presence of S100B in children with JIA provide evidence of BBB disruption and CNS involvement, particularly in those with higher disease activity. These findings underscore the importance of considering CNS health in the comprehensive management of JIA. Further research is required to elucidate the mechanisms behind CNS engagement in JIA and to develop treatments that address both joint and CNS manifestations of the disease.
Collapse
Affiliation(s)
- Lillemor Berntson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| | - Andreas Elfving
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | | | - Anders Öman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Fariborz Mobarrez
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [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] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
7
|
Faizan M, Sachan N, Verma O, Sarkar A, Rawat N, Pratap Singh M. Cerebrospinal fluid protein biomarkers in Parkinson's disease. Clin Chim Acta 2024; 556:117848. [PMID: 38417781 DOI: 10.1016/j.cca.2024.117848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Proteomic profiling is an effective way to identify biomarkers for Parkinson's disease (PD). Cerebrospinal fluid (CSF) has direct connectivity with the brain and could be a source of finding biomarkers and their clinical implications. Comparative proteomic profiling has shown that a group of differentially displayed proteins exist. The studies performed using conventional and classical tools also supported the occurrence of these proteins. Many studies have highlighted the potential of CSF proteomic profiling for biomarker identification and their clinical applications. Some of these proteins are useful for disease diagnosis and prediction. Proteomic profiling of CSF also has immense potential to distinguish PD from similar neurodegenerative disorders. A few protein biomarkers help in fundamental knowledge generation and clinical interpretation. However, the specific biomarker of PD is not yet known. The use of proteomic approaches in clinical settings is also rare. A large-scale, multi-centric, multi-population and multi-continental study using multiple proteomic tools is warranted. Such a study can provide valuable, comprehensive and reliable information for a better understanding of PD and the development of specific biomarkers. The current article sheds light on the role of CSF proteomic profiling in identifying biomarkers of PD and their clinical implications. The article also explains the achievements, obstacles and hopes for future directions of this approach.
Collapse
Affiliation(s)
- Mohd Faizan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Nidhi Sachan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Oyashvi Verma
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Alika Sarkar
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Neeraj Rawat
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Capacity Building and Knowledge Services, ASSIST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.
| |
Collapse
|
8
|
Nunes MJ, Carvalho AN, Rosa AI, Videira PA, Gama MJ, Rodrigues E, Castro-Caldas M. Altered expression of Sialyl Lewis X in experimental models of Parkinson's disease. J Mol Med (Berl) 2024; 102:365-377. [PMID: 38197965 PMCID: PMC10879467 DOI: 10.1007/s00109-023-02415-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
The mechanisms underlying neurodegeneration in Parkinson's disease (PD) are still not fully understood. Glycosylation is an important post-translational modification that affects protein function, cell-cell contacts and inflammation and can be modified in pathologic conditions. Although the involvement of aberrant glycosylation has been proposed for PD, the knowledge of the diversity of glycans and their role in PD is still minimal. Sialyl Lewis X (sLeX) is a sialylated and fucosylated tetrasaccharide with essential roles in cell-to-cell recognition processes. Pathological conditions and pro-inflammatory mediators can up-regulate sLeX expression on cell surfaces, which has important consequences in intracellular signalling and immune function. Here, we investigated the expression of this glycan using in vivo and in vitro models of PD. We show the activation of deleterious glycation-related pathways in mouse striatum upon treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a toxin-based model of PD. Importantly, our results show that MPTP triggers the presentation of more proteins decorated with sLeX in mouse cortex and striatum in a time-dependent manner, as well as increased mRNA expression of its rate-limiting enzyme fucosyltransferase 7. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. Although the underlying mechanism that drives increased sLeX epitopes, the nature of the protein scaffolds and their functional importance in PD remain unknown, our data suggest for the first time that sLeX in the brain may have a role in neuronal signalling and immunomodulation in pathological conditions. KEY MESSAGES: MPTP triggers the presentation of proteins decorated with sLeX in mouse brain. MPTP triggers the expression of sLeX rate-limiting enzyme FUT 7 in striatum. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. sLeX in the brain may have a role in neuronal signalling and immunomodulation.
Collapse
Affiliation(s)
- Maria João Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Alexandra I Rosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Paula A Videira
- Department of Life Sciences, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| | - Maria João Gama
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Elsa Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Margarida Castro-Caldas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
- Department of Life Sciences, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| |
Collapse
|
9
|
Garcia R, Zarate S, Srinivasan R. The Role of Astrocytes in Parkinson's Disease : Astrocytes in Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2024; 39:319-343. [PMID: 39190081 DOI: 10.1007/978-3-031-64839-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with a complex and multifactorial pathogenesis. This chapter delves into the critical role of astrocytes in PD. Once viewed as supporting cells in the central nervous system, astrocytes have emerged as key players in both maintaining neuronal health and contributing to neurodegeneration in PD. Their functions play a dual role in the progression of PD, ranging from protective functions like secretion of neurotrophic factors and clearance of α-synuclein to detrimental functions like promotion of neuroinflammation. This chapter is structured into three primary sections: the morphological and functional organization of astrocytes, astrocytic calcium signaling, and the role of astrocyte heterogeneity in PD. We provide a detailed exploration of astrocytic organelles, bidirectional astrocyte-neuron interactions, and the impact of astrocytic secretions such as antioxidant molecules and neurotrophic factors. Furthermore, we discuss the influence of astrocytes on non-neuronal cells, including interactions with microglia and the blood-brain barrier (BBB). By examining the multifaceted roles of astrocytes, in this chapter, we aim to bridge basic astrocyte biology with the clinical complexities of PD, offering insights into novel therapeutic strategies. The inclusion of astrocyte biology in our broader research approach will aid in the development of more effective treatment strategies for PD.
Collapse
Affiliation(s)
- Roger Garcia
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Sara Zarate
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
| |
Collapse
|
10
|
Yilmaz M, Yay E, Balci N, Toygar H, Kılıc BB, Zirh A, Rivas CA, Kantarci A. Parkinson's disease is positively associated with periodontal inflammation. J Periodontol 2023; 94:1425-1435. [PMID: 37433175 DOI: 10.1002/jper.23-0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Parkinson's disease (PA) affects 1% of the global population above 60 years. PA pathogenesis involves severe neuroinflammation that impacts systemic and local inflammatory changes. We tested the hypothesis that PA is associated with periodontal tissue inflammation promoting a greater systemic inflammatory burden. METHODS We recruited 60 patients with Stage III, Grade B periodontitis (P) with and without PA (n = 20 for each). We also included systemically and periodontally healthy individuals as controls (n = 20). Clinical periodontal parameters were recorded. Serum, saliva, and gingival crevicular fluid (GCF) samples were collected to measure the inflammatory and neurodegenerative targets (YKL-40, fractalkine, S100B, alpha-synuclein, tau, vascular cell adhesion protein-1 (VCAM-1), brain-derived neurotrophic factor (BDNF), neurofilament light chain (NfL). RESULTS Parkinson's patients in this study had mild to moderate motor dysfunctions, which did not prevent them from performing optimal oral hygiene control. Periodontal parameters and GCF volume were significantly higher in the P and P+PA groups than in the control group. PA was associated with significantly increased bleeding on probing (BOP) compared to P-alone (p < 0.05), while other clinical parameters were similar between P and P+PA groups. In saliva and serum, YKL-40 levels were higher in the P+PA group than in P and C groups (p < 0.001). GCF NfL levels from shallow sites were significantly higher in the P+PA group compared to the C group (p = 0.0462). GCF S100B levels from deep sites were higher in the P+PA group than in healthy individuals (p = 0.0194). CONCLUSION The data suggested that PA is highly associated with increased periodontal inflammatory burden-bleeding upon probing and inflammatory markers-in parallel with PA-related neuroinflammation.
Collapse
Affiliation(s)
- Melis Yilmaz
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Medipol University, Faculty of Dentistry, Department of Periodontology, Istanbul, Turkey
| | - Ekin Yay
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Medipol University, Faculty of Dentistry, Department of Periodontology, Istanbul, Turkey
| | - Nur Balci
- Medipol University, Faculty of Dentistry, Department of Periodontology, Istanbul, Turkey
| | - Hilal Toygar
- Medipol University, Faculty of Dentistry, Department of Periodontology, Istanbul, Turkey
| | - Basak Bolluk Kılıc
- Medipol University, Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Ali Zirh
- Medipol University, Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Carla Alvarez Rivas
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Alpdogan Kantarci
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Harvard School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Jiang G, Sheng C, Yan L, Wang Z, Wang Q, Chen R, Zhao Y. Increased Serum S100β Concentration is Associated with Depression in Parkinson's Disease. Neuropsychiatr Dis Treat 2023; 19:1865-1873. [PMID: 37663392 PMCID: PMC10473424 DOI: 10.2147/ndt.s423312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023] Open
Abstract
Purpose To explore the relationship between the serum level of S100 calcium-binding protein, beta chain (S100β) and Parkinson's disease (PD) with depression. Patients and Methods A total of 145 patients with PD and 60 healthy controls matched for sex, age, and years of education in our hospital were selected. Fluorescence quantitative immunochromatography was used to quantify the level of S100β in serum. Clinical manifestations were assessed by Unified Parkinson's Disease Rating Scale part-III (UPDRS-III), Hoehn & Yahr (H-Y) stage and 17-item Hamilton Rating Scale for Depression (HAMD-17). According to the results of HAMD-17, PD patients were divided into PD with depression group and PD without depression group. The relationship between serum S100β and HAMD-17 scores in PD patients with depression was investigated through correlation analysis and multivariate regression analysis, and receiver operating characteristic (ROC) curve was used to evaluate the diagnostic value of serum S100β. Results The level of serum S100β in PD with depression group was significantly higher than that in PD without depression group and control group. In PD patients with depression, serum S100β level was positively correlated with UPDRS-III score, H-Y Scale and HAMD-17 score. The HAMD-17 score was positively correlated with the UPDRS-III and H-Y scales, and the increase in the HAMD-17 score was associated with women. Elevated serum S100β level and UPDRS-III score are independent risk factors for PD with depression. Analysis of receiver operating characteristic (ROC) curves showed that the serum S100β level with a cutoff of 0.28 ng/mL distinguished patients with PD with or without depression with an area under the ROC curve (AUC) of 0.742, sensitivity of 0.696, and specificity of 0.779. Conclusion The serum S100β level could be a biomarker of PD with depression.
Collapse
Affiliation(s)
- Guanghui Jiang
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Cuicui Sheng
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Luxia Yan
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Zipeng Wang
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Qing Wang
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Rui Chen
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| | - Ying Zhao
- Department of Neurology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, People’s Republic of China
| |
Collapse
|
12
|
Giovannini G, Meletti S. Fluid Biomarkers of Neuro-Glial Injury in Human Status Epilepticus: A Systematic Review. Int J Mol Sci 2023; 24:12519. [PMID: 37569895 PMCID: PMC10420319 DOI: 10.3390/ijms241512519] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
As per the latest ILAE definition, status epilepticus (SE) may lead to long-term irreversible consequences, such as neuronal death, neuronal injury, and alterations in neuronal networks. Consequently, there is growing interest in identifying biomarkers that can demonstrate and quantify the extent of neuronal and glial injury. Despite numerous studies conducted on animal models of status epilepticus, which clearly indicate seizure-induced neuronal and glial injury, as well as signs of atrophy and gliosis, evidence in humans remains limited to case reports and small case series. The implications of identifying such biomarkers in clinical practice are significant, including improved prognostic stratification of patients and the early identification of those at high risk of developing irreversible complications. Moreover, the clinical validation of these biomarkers could be crucial in promoting neuroprotective strategies in addition to antiseizure medications. In this study, we present a systematic review of research on biomarkers of neuro-glial injury in patients with status epilepticus.
Collapse
Affiliation(s)
- Giada Giovannini
- Neurology Department, Azienda Ospedaliera-Universitaria di Modena, 41126 Modena, Italy;
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio-Emilia, 41121 Modena, Italy
| | - Stefano Meletti
- Neurology Department, Azienda Ospedaliera-Universitaria di Modena, 41126 Modena, Italy;
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio-Emilia, 41121 Modena, Italy
| |
Collapse
|
13
|
Chang NP, DaPrano EM, Evans WR, Nissenbaum M, McCourt M, Alzate D, Lindman M, Chou TW, Atkins C, Kusnecov AW, Huda R, Daniels BP. Neuronal DAMPs exacerbate neurodegeneration via astrocytic RIPK3 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550097. [PMID: 37546744 PMCID: PMC10401942 DOI: 10.1101/2023.07.21.550097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Astrocyte activation is a common feature of neurodegenerative diseases. However, the ways in which dying neurons influence the activity of astrocytes is poorly understood. RIPK3 signaling has recently been described as a key regulator of neuroinflammation, but whether this kinase mediates astrocytic responsiveness to neuronal death has not yet been studied. Here, we used the MPTP model of Parkinson's disease to show that activation of astrocytic RIPK3 drives dopaminergic cell death and axon damage. Transcriptomic profiling revealed that astrocytic RIPK3 promoted gene expression associated with neuroinflammation and movement disorders, and this coincided with significant engagement of DAMP signaling. Using human cell culture systems, we show that factors released from dying neurons signal through RAGE to induce RIPK3-dependent astrocyte activation. These findings highlight a mechanism of neuron-glia crosstalk in which neuronal death perpetuates further neurodegeneration by engaging inflammatory astrocyte activation via RIPK3.
Collapse
Affiliation(s)
- Nydia P. Chang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Evan M. DaPrano
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Wesley R. Evans
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Diego Alzate
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Rafiq Huda
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
14
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 191] [Impact Index Per Article: 191.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
15
|
Michetti F, Clementi ME, Di Liddo R, Valeriani F, Ria F, Rende M, Di Sante G, Romano Spica V. The S100B Protein: A Multifaceted Pathogenic Factor More Than a Biomarker. Int J Mol Sci 2023; 24:ijms24119605. [PMID: 37298554 DOI: 10.3390/ijms24119605] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
S100B is a calcium-binding protein mainly concentrated in astrocytes in the nervous system. Its levels in biological fluids are recognized as a reliable biomarker of active neural distress, and more recently, mounting evidence points to S100B as a Damage-Associated Molecular Pattern molecule, which, at high concentration, triggers tissue reactions to damage. S100B levels and/or distribution in the nervous tissue of patients and/or experimental models of different neural disorders, for which the protein is used as a biomarker, are directly related to the progress of the disease. In addition, in experimental models of diseases such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, multiple sclerosis, traumatic and vascular acute neural injury, epilepsy, and inflammatory bowel disease, alteration of S100B levels correlates with the occurrence of clinical and/or toxic parameters. In general, overexpression/administration of S100B worsens the clinical presentation, whereas deletion/inactivation of the protein contributes to the amelioration of the symptoms. Thus, the S100B protein may be proposed as a common pathogenic factor in different disorders, sharing different symptoms and etiologies but appearing to share some common pathogenic processes reasonably attributable to neuroinflammation.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Department of Neuroscience, Catholic University of the Sacred Heart, 00168 Rome, Italy
- IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Department of Medicine, LUM University, 70010 Casamassima, Italy
- Genes, Via Venti Settembre 118, 00187 Roma, Italy
| | | | - Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Federica Valeriani
- Laboratory of Epidemiology and Biotechnologies, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", 00135 Rome, Italy
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06132 Perugia, Italy
| | - Gabriele Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06132 Perugia, Italy
| | - Vincenzo Romano Spica
- Laboratory of Epidemiology and Biotechnologies, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", 00135 Rome, Italy
| |
Collapse
|
16
|
Leem YH, Kim DY, Park JE, Kim HS. Necrosulfonamide exerts neuroprotective effect by inhibiting necroptosis, neuroinflammation, and α-synuclein oligomerization in a subacute MPTP mouse model of Parkinson's disease. Sci Rep 2023; 13:8783. [PMID: 37258791 DOI: 10.1038/s41598-023-35975-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023] Open
Abstract
Parkinson's disease (PD) is an incurable movement disorder characterized by dopaminergic cell loss, neuroinflammation, and α-synuclein pathology. Herein, we investigated the therapeutic effects of necrosulfonamide (NSA), a specific inhibitor of mixed lineage kinase domain-like protein (MLKL), in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. MLKL is an executor of necroptosis, a programmed cell death pathway that causes inflammation. Repeated administration of NSA resulted in the recovery of impaired motor performance and dopaminergic degeneration. Furthermore, NSA inhibited the phosphorylation, ubiquitylation, and oligomerization of MLKL, all of which are associated with MLKL cell death-inducing activity in dopaminergic cells in the substantia nigra (SN). NSA also inhibited microglial activation and reactive astrogliosis as well as the MPTP-induced expression of proinflammatory molecules such as tumor necrosis factor-α, interleukin-1β, inducible nitric oxide synthase, and cystatin F. Furthermore, NSA inhibited α-synuclein oligomerization and phosphorylation in the SN of MPTP-treated mice by inhibiting the activity of glycogen synthase kinase 3β and matrix metalloproteinase-3. In conclusion, NSA has anti-necroptotic, anti-inflammatory, and anti-synucleinopathic effects on PD pathology. Therefore, NSA is a potential therapeutic candidate for PD.
Collapse
Affiliation(s)
- Yea-Hyun Leem
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-Dong, Gangseo-gu, Seoul, 07804, South Korea.
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
17
|
Bourque M, Morissette M, Soulet D, Di Paolo T. Impact of Sex on Neuroimmune contributions to Parkinson's disease. Brain Res Bull 2023:110668. [PMID: 37196734 DOI: 10.1016/j.brainresbull.2023.110668] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/27/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. Inflammation has been observed in both the idiopathic and familial forms of PD. Importantly, PD is reported more often in men than in women, men having at least 1.5- fold higher risk to develop PD than women. This review summarizes the impact of biological sex and sex hormones on the neuroimmune contributions to PD and its investigation in animal models of PD. Innate and peripheral immune systems participate in the brain neuroinflammation of PD patients and is reproduced in neurotoxin, genetic and alpha-synuclein based models of PD. Microglia and astrocytes are the main cells of the innate immune system in the central nervous system and are the first to react to restore homeostasis in the brain. Analysis of serum immunoprofiles in female and male control and PD patients show that a great proportion of these markers differ between male and female. The relationship between CSF inflammatory markers and PD clinical characteristics or PD biomarkers shows sex differences. Conversely, in animal models of PD, sex differences in inflammation are well documented and the beneficial effects of endogenous and exogenous estrogenic modulation in inflammation have been reported. Targeting neuroinflammation in PD is an emerging therapeutic option but gonadal drugs have not yet been investigated in this respect, thus offering new opportunities for sex specific treatments.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| |
Collapse
|
18
|
Lillian A, Zuo W, Laham L, Hilfiker S, Ye JH. Pathophysiology and Neuroimmune Interactions Underlying Parkinson's Disease and Traumatic Brain Injury. Int J Mol Sci 2023; 24:7186. [PMID: 37108349 PMCID: PMC10138999 DOI: 10.3390/ijms24087186] [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: 02/07/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder clinically defined by motor instability, bradykinesia, and resting tremors. The clinical symptomatology is seen alongside pathologic changes, most notably the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of α-synuclein and neuromelanin aggregates throughout numerous neural circuits. Traumatic brain injury (TBI) has been implicated as a risk factor for developing various neurodegenerative diseases, with the most compelling argument for the development of PD. Dopaminergic abnormalities, the accumulation of α-synuclein, and disruptions in neural homeostatic mechanisms, including but not limited to the release of pro-inflammatory mediators and the production of reactive oxygen species (ROS), are all present following TBI and are closely related to the pathologic changes seen in PD. Neuronal iron accumulation is discernable in degenerative and injured brain states, as is aquaporin-4 (APQ4). APQ4 is an essential mediator of synaptic plasticity in PD and regulates edematous states in the brain after TBI. Whether the cellular and parenchymal changes seen post-TBI directly cause neurodegenerative diseases such as PD is a point of considerable interest and debate; this review explores the vast array of neuroimmunological interactions and subsequent analogous changes that occur in TBI and PD. There is significant interest in exploring the validity of the relationship between TBI and PD, which is a focus of this review.
Collapse
Affiliation(s)
- Alyssa Lillian
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Wanhong Zuo
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Linda Laham
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Sabine Hilfiker
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| |
Collapse
|
19
|
Chegão A, Vicente Miranda H. Unveiling new secrets in Parkinson's disease: The glycatome. Behav Brain Res 2023; 442:114309. [PMID: 36706808 DOI: 10.1016/j.bbr.2023.114309] [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: 11/15/2022] [Revised: 01/04/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
We are witnessing a considerable increase in the incidence of Parkinson's disease (PD), which may be due to the general ageing of the population. While there is a plethora of therapeutic strategies for this disease, they still fail to arrest disease progression as they do not target and prevent the neurodegenerative process. The identification of disease-causing mutations allowed researchers to better dissect the underlying causes of this disease, highlighting, for example, the pathogenic role of alpha-synuclein. However, most PD cases are sporadic, which is making it hard to unveil the major causative mechanisms of this disease. In the recent years, epidemiological evidence suggest that type-2 diabetes mellitus (T2DM) individuals have higher risk and worst outcomes of PD, allowing to raise the hypothesis that some dysregulated processes in T2DM may contribute or even trigger the neurodegenerative process in PD. One major consequence of T2DM is the unprogrammed reaction between sugars, increased in T2DM, and proteins, a reaction named glycation. Pre-clinical reports show that alpha-synuclein is a target of glycation, and glycation potentiates its pathogenicity which contributes for the neurodegenerative process. Moreover, it triggers, anticipates, or aggravates several PD-like motor and non-motor complications. A given profile of proteins are differently glycated in diseased conditions, altering the brain proteome and leading to brain dysfunction and neurodegeneration. Herein we coin the term Glycatome as the profile of glycated proteins. In this review we report on the mechanisms underlying the association between T2DM and PD, with particular focus on the impact of protein glycation.
Collapse
Affiliation(s)
- Ana Chegão
- iNOVA4Health, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hugo Vicente Miranda
- iNOVA4Health, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisboa, Portugal.
| |
Collapse
|
20
|
Intracellular DAMPs in Neurodegeneration and Their Role in Clinical Therapeutics. Mol Neurobiol 2023; 60:3600-3616. [PMID: 36859688 DOI: 10.1007/s12035-023-03289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023]
Abstract
Neuroinflammation is the major implication of neurodegeneration. This is a complex process which initiates from the cellular injury triggering the innate immune system which gives rise to damage-associated molecular patterns (DAMPs) which are also recognized as endogenous danger indicators. These originate from various compartments of the cell under pathological stimulus. These are very popular candidates having their origin in the intracellular compartments and organelles of the cell and may have their site of action itself in the intracellular or at the extracellular spaces. Under the influence of the pathological stimuli, they interact with the pattern-recognition receptor to initiate their pro-inflammatory cascade followed by the cytokine release. This provides a good opportunity for diagnostic and therapeutic interventions creating better conditions for repair and reversal. Since the major contributors arise from the intracellular compartment, in this review, we have attempted to focus on the DAMP molecules arising from the intracellular compartments and their specific roles in the neurodegenerative events explaining their downstream mediators and signaling. Moreover, we have tried to cover the latest interventions in terms of DAMPs as clinical biomarkers which can assist in detecting the disease and also target it to reduce the innate-immune activation response which can help in reducing the sterile neuroinflammation having an integral role in the neurodegenerative processes.
Collapse
|
21
|
Kartik S, Pal R, Chaudhary MJ, Tiwari PC, Nath R, Kumar M. Anti-oxidative and anti-neuroinflammatory role of Necrostatin-1s and docosahexaenoic acid in RIP-1-mediated neurotoxicity in MPTP-induced Parkinson's disease model. Fundam Clin Pharmacol 2023. [PMID: 36807936 DOI: 10.1111/fcp.12881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/17/2023] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
Parkinson's disease (PD) is a neuromuscular ailment that affects people in their later years and causes both motor and non-motor deficits. Receptor-interacting protein-1 (RIP-1) is a critical participant in necroptotic cell death, possibly through an oxidant-antioxidant imbalance and cytokine cascade activation in PD pathogenesis. The present study examined the role of RIP-1-mediated necroptosis and neuroinflammation in the MPTP-induced PD mouse model, as well as their protection by Necrostatin-1s (an RIP signalling inhibitor), antioxidant DHA and their functional interaction. BALB/c mice were given acute MPTP therapy (4 injections of 15 mg/kg i.p. at 2-h intervals) on day 1. After MPTP intoxication, Necrostatin-1s (Nec-1s; 8 mg/kg/day, i.p.) and DHA (300 mg/kg/day, p.o.) treatments were given once daily for 7 days. The Nec-1s treatment prevented MPTP-induced behavioural, biochemical and neurochemical alterations, and the addition of DHA increases Nec-1s' neuroprotective impact. In addition, Nec-1s and DHA significantly improve the survival of TH-positive dopaminergic neurons and lower expression levels of the inflammatory cytokines, IL-1β and TNF-α. Furthermore, Nec-1s dramatically reduced RIP-1 expression, whereas DHA had little effect. Our research raises the possibility that neuroinflammatory signalling and acute MPTP-induced necroptosis are both mediated by TNFR1-driven RIP-1 activity. In this study, RIP-1 ablation through Nec-1s and the addition of DHA showed a reduction in the levels of pro-inflammatory and oxidative markers, as well as protection from MPTP-driven dopaminergic degeneration and neurobehavioural changes, suggesting potential therapeutic applications. For a better understanding, additional research about the mechanism(s) behind Nec-1s and DHA is required.
Collapse
Affiliation(s)
- Shipra Kartik
- Department of Pharmacology & Therapeutics, King George's Medical University, 226003, Lucknow, India
| | - Rishi Pal
- Department of Pharmacology & Therapeutics, King George's Medical University, 226003, Lucknow, India
| | - Manju J Chaudhary
- Department of Physiology, Government Medical College, Tirwa Road, Kannauj, India
| | - Prafulla Chandra Tiwari
- Department of Pharmacology & Therapeutics, King George's Medical University, 226003, Lucknow, India
| | - Rajendra Nath
- Department of Pharmacology & Therapeutics, King George's Medical University, 226003, Lucknow, India
| | - Madhu Kumar
- Department of Pathology, King George's Medical University, Lucknow, 226003, India
| |
Collapse
|
22
|
Wang Q, Zheng J, Pettersson S, Reynolds R, Tan EK. The link between neuroinflammation and the neurovascular unit in synucleinopathies. SCIENCE ADVANCES 2023; 9:eabq1141. [PMID: 36791205 PMCID: PMC9931221 DOI: 10.1126/sciadv.abq1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 01/19/2023] [Indexed: 05/28/2023]
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Karolinska Institutet, Department of Odontology, 171 77 Solna, Sweden
- Faculty of Medical Sciences, Sunway University, Subang Jaya, 47500 Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
23
|
Cullinane PW, de Pablo Fernandez E, König A, Outeiro TF, Jaunmuktane Z, Warner TT. Type 2 Diabetes and Parkinson's Disease: A Focused Review of Current Concepts. Mov Disord 2023; 38:162-177. [PMID: 36567671 DOI: 10.1002/mds.29298] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 12/27/2022] Open
Abstract
Highly reproducible epidemiological evidence shows that type 2 diabetes (T2D) increases the risk and rate of progression of Parkinson's disease (PD), and crucially, the repurposing of certain antidiabetic medications for the treatment of PD has shown early promise in clinical trials, suggesting that the effects of T2D on PD pathogenesis may be modifiable. The high prevalence of T2D means that a significant proportion of patients with PD may benefit from personalized antidiabetic treatment approaches that also confer neuroprotective benefits. Therefore, there is an immediate need to better understand the mechanistic relation between these conditions and the specific molecular pathways affected by T2D in the brain. Although there is considerable evidence that processes such as insulin signaling, mitochondrial function, autophagy, and inflammation are involved in the pathogenesis of both PD and T2D, the primary aim of this review is to highlight the evidence showing that T2D-associated dysregulation of these pathways occurs not only in the periphery but also in the brain and how this may facilitate neurodegeneration in PD. We also discuss the challenges involved in disentangling the complex relationship between T2D, insulin resistance, and PD, as well as important questions for further research. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eduardo de Pablo Fernandez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
24
|
Is S100B Involved in Attention-Deficit/Hyperactivity Disorder (ADHD)? Comparisons with Controls and Changes Following a Triple Therapy Containing Methylphenidate, Melatonin and ω-3 PUFAs. Nutrients 2023; 15:nu15030712. [PMID: 36771418 PMCID: PMC9919946 DOI: 10.3390/nu15030712] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Increasing evidence supports a neuroinflammatory basis in ADHD damaging glial function and thereby altering dopaminergic (DA) neurotransmission. Previous studies focusing on the S100B protein as a marker of glial function have shown contradictory results. We conducted a clinical trial to investigate differences in S100B levels between ADHD patients and controls, as well as observe gradual changes in S100B concentrations after a triple therapy (TT) containing methylphenidate (MPH), melatonin (aMT) and omega-3 fatty acids (ω-3 PUFAs). METHODS 62 medication-naïve children with ADHD (ADHD-G) and 65 healthy controls (C-G) were recruited. Serum S100B was measured at baseline (T0) in ADHD-G/C-G, and three (T3) and six months (T6) after starting TT in the ADHD-G, together with attention scores. RESULTS A significant increase in S100B was observed in the ADHD-G vs. C-G. In the ADHD-G, significantly higher S100B values were observed for comparisons between T0-T3 and between T0-T6, accompanied by a significant improvement in attention scores for the same timepoint comparisons. No significant differences were found for S100B between T3-T6. CONCLUSION Our results agree with the hypothesis of glial damage in ADHD. Further studies on the link between DA and S100B are required to explain the transient increase in S100B following TT.
Collapse
|
25
|
Rossi R, Douglas A, Gil SM, Jabrah D, Pandit A, Gilvarry M, McCarthy R, Prendergast J, Jood K, Redfors P, Nordanstig A, Ceder E, Dunker D, Carlqvist J, Szikora I, Thornton J, Tsivgoulis G, Psychogios K, Tatlisumak T, Rentzos A, Doyle KM. S100b in acute ischemic stroke clots is a biomarker for post-thrombectomy intracranial hemorrhages. Front Neurol 2023; 13:1067215. [PMID: 36756347 PMCID: PMC9900124 DOI: 10.3389/fneur.2022.1067215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/01/2022] [Indexed: 01/24/2023] Open
Abstract
Background and purpose Post-thrombectomy intracranial hemorrhages (PTIH) are dangerous complications of acute ischemic stroke (AIS) following mechanical thrombectomy. We aimed to investigate if S100b levels in AIS clots removed by mechanical thrombectomy correlated to increased risk of PTIH. Methods We analyzed 122 thrombi from 80 AIS patients in the RESTORE Registry of AIS clots, selecting an equal number of patients having been pre-treated or not with rtPA (40 each group). Within each subgroup, 20 patients had developed PTIH and 20 patients showed no signs of hemorrhage. Gross photos of each clot were taken and extracted clot area (ECA) was measured using ImageJ. Immunohistochemistry for S100b was performed and Orbit Image Analysis was used for quantification. Immunofluorescence was performed to investigate co-localization between S100b and T-lymphocytes, neutrophils and macrophages. Chi-square or Kruskal-Wallis test were used for statistical analysis. Results PTIH was associated with higher S100b levels in clots (0.33 [0.08-0.85] vs. 0.07 [0.02-0.27] mm2, H1 = 6.021, P = 0.014*), but S100b levels were not significantly affected by acute thrombolytic treatment (P = 0.386). PTIH was also associated with patients having higher NIHSS at admission (20.0 [17.0-23.0] vs. 14.0 [10.5-19.0], H1 = 8.006, P = 0.005) and higher number of passes during thrombectomy (2 [1-4] vs. 1 [1-2.5], H1 = 5.995, P = 0.014*). S100b co-localized with neutrophils, macrophages and with T-lymphocytes in the clots. Conclusions Higher S100b expression in AIS clots, higher NIHSS at admission and higher number of passes during thrombectomy are all associated with PTIH. Further investigation of S100b expression in AIS clots by neutrophils, macrophages and T-lymphocytes could provide insight into the role of S100b in thromboinflammation.
Collapse
Affiliation(s)
- Rosanna Rossi
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland,*Correspondence: Rosanna Rossi ✉
| | - Andrew Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Sara Molina Gil
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Duaa Jabrah
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | | | - James Prendergast
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Katarina Jood
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Redfors
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Annika Nordanstig
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Erik Ceder
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Dennis Dunker
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Jeanette Carlqvist
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - István Szikora
- Department of Neurointerventions, National Institute of Clinical Neurosciences, Budapest, Hungary
| | - John Thornton
- Department of Radiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Turgut Tatlisumak
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alexandros Rentzos
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Karen M. Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland,Karen M. Doyle ✉
| |
Collapse
|
26
|
Hu J, Xie S, Li W, Zhang L. Diagnostic and prognostic value of serum S100B in sepsis-associated encephalopathy: A systematic review and meta-analysis. Front Immunol 2023; 14:1102126. [PMID: 36776893 PMCID: PMC9911439 DOI: 10.3389/fimmu.2023.1102126] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Background In sepsis, brain dysfunction is known as Sepsis-associated encephalopathy (SAE), which often results in severe cognitive and neurological sequelae and increases the risk of death. Our systematic review and meta-analysis aimed to explore the diagnostic and prognostic value of serum S100 calcium-binding protein B (S100B) in SAE patients. Methods We conducted a systematic search of the databases PubMed, Web of Science, Embase, Cochrane databases, CNKI, VIP, and WFSD from their inception dates until August 20, 2022. A Meta-analysis of the included studies was also performed using Review Manager version 5.4 and Stata16.0. Results This meta-analysis included 28 studies with 1401 serum samples from SAE patients and 1591 serum samples from no-encephalopathy septic (NE) patients. The Meta-Analysis showed that individuals with SAE had higher serum S100B level than NE controls (MD, 0.49 [95% CI (0.37)-(0.60), Z =8.29, P < 0.00001]), and the baseline level of serum S100B in septic patients with burn was significantly higher than average (1.96 [95% CI (0.92)-(2.99), Z =3.71, P < 0.0002]) In addition, septic patients with favorable outcomes had lower serum S100B levels than those with unfavorable outcomes (MD, -0.35 [95% CI (-0.50)-(-0.20), Z =4.60, P < 0.00001]). Conclusion Our Meta-Analysis indicates that higher serum S100B level in septic patients are moderately associated with SAE and unfavorable outcomes (The outcomes here mainly refer to the mortality). The serum S100B level may be a useful diagnostic and prognostic biomarker of SAE.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shucai Xie
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenchao Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
27
|
Koerich S, Parreira GM, de Almeida DL, Vieira RP, de Oliveira ACP. Receptors for Advanced Glycation End Products (RAGE): Promising Targets Aiming at the Treatment of Neurodegenerative Conditions. Curr Neuropharmacol 2023; 21:219-234. [PMID: 36154605 PMCID: PMC10190138 DOI: 10.2174/1570159x20666220922153903] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
Advanced glycation end products (AGEs) are compounds formed after the non-enzymatic addition of reducing sugars to lipids, proteins, and nucleic acids. They are associated with the development of various clinical complications observed in diabetes and cardiovascular diseases, such as retinopathy, nephropathy, diabetic neuropathy, and others. In addition, compelling evidence indicates that these molecules participate in the progression of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Multiple cellular and molecular alterations triggered by AGEs that could alter homeostasis have been identified. One of the main targets for AGE signaling is the receptor for advanced glycation end-products (RAGE). Importantly, this receptor is the target of not only AGEs, but also amyloid β peptides, HMGB1 (high-mobility group box-1), members of the S100 protein family, and glycosaminoglycans. The activation of this receptor induces intracellular signaling cascades that are involved in pathological processes and cell death. Therefore, RAGE represents a key target for pharmacological interventions in neurodegenerative diseases. This review will discuss the various effects of AGEs and RAGE activation in the pathophysiology of neurodegenerative diseases, as well as the currently available pharmacological tools and promising drug candidates.
Collapse
Affiliation(s)
- Suélyn Koerich
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Gabriela Machado Parreira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Rafael Pinto Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | |
Collapse
|
28
|
Brown TG, Thayer MN, VanTreeck JG, Zarate N, Hart DW, Heilbronner S, Gomez-Pastor R. Striatal spatial heterogeneity, clustering, and white matter association of GFAP + astrocytes in a mouse model of Huntington's disease. Front Cell Neurosci 2023; 17:1094503. [PMID: 37187609 PMCID: PMC10175581 DOI: 10.3389/fncel.2023.1094503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction Huntington's disease (HD) is a neurodegenerative disease that primarily affects the striatum, a brain region that controls movement and some forms of cognition. Neuronal dysfunction and loss in HD is accompanied by increased astrocyte density and astrocyte pathology. Astrocytes are a heterogeneous population classified into multiple subtypes depending on the expression of different gene markers. Studying whether mutant Huntingtin (HTT) alters specific subtypes of astrocytes is necessary to understand their relative contribution to HD. Methods Here, we studied whether astrocytes expressing two different markers; glial fibrillary acidic protein (GFAP), associated with astrocyte activation, and S100 calcium-binding protein B (S100B), a marker of matured astrocytes and inflammation, were differentially altered in HD. Results First, we found three distinct populations in the striatum of WT and symptomatic zQ175 mice: GFAP+, S100B+, and dual GFAP+S100B+. The number of GFAP+ and S100B+ astrocytes throughout the striatum was increased in HD mice compared to WT, coinciding with an increase in HTT aggregation. Overlap between GFAP and S100B staining was expected, but dual GFAP+S100B+ astrocytes only accounted for less than 10% of all tested astrocytes and the number of GFAP+S100B+ astrocytes did not differ between WT and HD, suggesting that GFAP+ astrocytes and S100B+ astrocytes are distinct types of astrocytes. Interestingly, a spatial characterization of these astrocyte subtypes in HD mice showed that while S100B+ were homogeneously distributed throughout the striatum, GFAP+ preferentially accumulated in "patches" in the dorsomedial (dm) striatum, a region associated with goal-directed behaviors. In addition, GFAP+ astrocytes in the dm striatum of zQ175 mice showed increased clustering and association with white matter fascicles and were preferentially located in areas with low HTT aggregate load. Discussion In summary, we showed that GFAP+ and S100B+ astrocyte subtypes are distinctly affected in HD and exist in distinct spatial arrangements that may offer new insights to the function of these specific astrocytes subtypes and their potential implications in HD pathology.
Collapse
|
29
|
Gelain DP, Bittencourt RR, Bastos Mendes LF, Moreira JCF, Outeiro TF. RAGE Against the Glycation Machine in Synucleinopathies: Time to Explore New Questions. JOURNAL OF PARKINSON'S DISEASE 2023; 13:717-728. [PMID: 37270812 PMCID: PMC10473104 DOI: 10.3233/jpd-230070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Oligomerization and aggregation of misfolded forms of α-synuclein are believed to be key molecular mechanisms in Parkinson's disease (PD) and other synucleinopathies, so extensive research has attempted to understand these processes. Among diverse post-translational modifications that impact α-synuclein aggregation, glycation may take place at several lysine sites and modify α-synuclein oligomerization, toxicity, and clearance. The receptor for advanced glycation end products (RAGE) is considered a key regulator of chronic neuroinflammation through microglial activation in response to advanced glycation end products, such as carboxy-ethyl-lysine, or carboxy-methyl-lysine. The presence of RAGE in the midbrain of PD patients has been reported in the last decades and this receptor was proposed to have a role in sustaining PD neuroinflammation. However, different PD animal models demonstrated that RAGE is preferentially expressed in neurons and astrocytes, while recent evidence demonstrated that fibrillar, non-glycated α-synuclein binds to RAGE. Here, we summarize the available data on α-synuclein glycation and RAGE in the context of PD, and discuss about the questions yet to be answered that may increase our understanding of the molecular bases of PD and synucleinopathies.
Collapse
Affiliation(s)
- Daniel Pens Gelain
- Center for Oxidative Stress Studies, Department of Biochemistry, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Reykla Ramon Bittencourt
- Center for Oxidative Stress Studies, Department of Biochemistry, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz Filipe Bastos Mendes
- Center for Oxidative Stress Studies, Department of Biochemistry, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - José Claudio Fonseca Moreira
- Center for Oxidative Stress Studies, Department of Biochemistry, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Natural Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| |
Collapse
|
30
|
Bancroft EA, De La Mora M, Pandey G, Zarate SM, Srinivasan R. Extracellular S100B inhibits A-type voltage-gated potassium currents and increases L-type voltage-gated calcium channel activity in dopaminergic neurons. Glia 2022; 70:2330-2347. [PMID: 35916350 PMCID: PMC10738449 DOI: 10.1002/glia.24254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022]
Abstract
Parkinson's disease (PD) is associated with an increase in secreted S100B within the midbrain and cerebrospinal fluid. In addition, S100B overexpression in mice accelerates the loss of substantia nigra pars compacta dopaminergic (DA) neurons, suggesting a role for this protein in PD pathogenesis. We found that in the mouse SNc, S100B labeled astrocytic processes completely envelop the somata of tyrosine hydroxylase (TH) expressing DA neurons only in male mice. These data suggest that an increase in S100B secretion by astrocytes within the midbrain could play a role in DA dysfunction during early PD. We therefore asked if acute exposure to extracellular S100B alters the activity of identified TH expressing DA neurons in primary mouse midbrain cultures. Acute exposure to 50 pM S100B specifically inhibited A-type voltage-gated potassium currents in TH+ , but not TH- neurons. This was accompanied by ~2-fold increases in the frequency of both intrinsic firing, as well as L-type voltage-gated calcium channel (VGCC)-mediated calcium fluxes only in TH+ neurons. Further, exposure to 100 μM 4-aminopyridine (4-AP), an A-type voltage-gated potassium channel inhibitor, mimicked the S100B mediated increase in intrinsic firing and L-type VGCC-mediated calcium fluxes in TH+ neurons. Taken together, our finding that extracellular S100B alters the activity of native DA neurons via an inhibition of A-type voltage-gated potassium channels has important implications for understanding the pathophysiology of early PD.
Collapse
Affiliation(s)
- Eric A. Bancroft
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas, USA
| | - Martha De La Mora
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas, USA
| | - Gauri Pandey
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience (TAMIN), College Station, Texas, USA
| | - Sara M. Zarate
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience (TAMIN), College Station, Texas, USA
| |
Collapse
|
31
|
Prunell G, Olivera-Bravo S. A Focus on Astrocyte Contribution to Parkinson's Disease Etiology. Biomolecules 2022; 12:biom12121745. [PMID: 36551173 PMCID: PMC9775515 DOI: 10.3390/biom12121745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disease of high prevalence, characterized by the prominent death of dopaminergic neurons in the substantia nigra pars compacta, which produces dopamine deficiency, leading to classic motor symptoms. Although PD has traditionally been considered as a neuronal cell autonomous pathology, in which the damage of vulnerable neurons is responsible for the disease, growing evidence strongly suggests that astrocytes might have an active role in the neurodegeneration observed. In the present review, we discuss several studies evidencing astrocyte implications in PD, highlighting the consequences of both the loss of normal homeostatic functions and the gain in toxic functions for the wellbeing of dopaminergic neurons. The revised information provides significant evidence that allows astrocytes to be positioned as crucial players in PD etiology, a factor that needs to be taken into account when considering therapeutic targets for the treatment of the disease.
Collapse
Affiliation(s)
- Giselle Prunell
- Laboratorio de Neurodegeneración y Neuroprotección, Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, Montevideo 11600, Uruguay
- Correspondence: (G.P.); (S.O.-B.); Tel.: +598-24871616 (ext. 121 or 123 or 171) (G.P. & S.O.-B.)
| | - Silvia Olivera-Bravo
- Laboratorio de Neurobiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, Montevideo 11600, Uruguay
- Correspondence: (G.P.); (S.O.-B.); Tel.: +598-24871616 (ext. 121 or 123 or 171) (G.P. & S.O.-B.)
| |
Collapse
|
32
|
Long H, Zhang S, Zeng S, Tong Y, Liu J, Liu C, Li D. Interaction of RAGE with α-synuclein fibrils mediates inflammatory response of microglia. Cell Rep 2022; 40:111401. [PMID: 36130498 DOI: 10.1016/j.celrep.2022.111401] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/03/2022] [Accepted: 08/31/2022] [Indexed: 11/03/2022] Open
Abstract
Microglia-mediated neuroinflammation and α-synuclein (α-syn) aggregation, both as pathological hallmarks of Parkinson's disease (PD), crosstalk to exacerbate degeneration of dopaminergic neurons and PD progression. However, the mechanism underlying their interaction is poorly understood, which obstructs effective therapeutic inhibition of α-syn-induced neuroinflammation. Here, we initiate from structure-based interaction predictions and find that receptor for advanced glycation end products (RAGE) serves as a receptor of α-syn fibrils on microglia. Results of nuclear magnetic resonance (NMR) spectroscopy and mutagenesis validate that the V domain of RAGE that contains an alkaline surface can bind with acidic C-terminal residues of α-syn. Furthermore, the binding of α-syn fibrils with RAGE induces neuroinflammation, which is blocked by both genetic depletion of RAGE and inhibitor FPS-ZM1. Our work shows the important role, as well as the structural mechanism, of RAGE in mediating the inflammatory response of microglia to α-syn fibrils, which may help to establish effective therapeutic strategies to alleviate α-syn-induced neuroinflammation and neuronal damage.
Collapse
Affiliation(s)
- Houfang Long
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyi Zeng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yilun Tong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China; WLA Laboratories, World Laureates Association, Shanghai 201203, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
33
|
Oh HN, Yoo D, Park S, Lee S, Kim WK. Developmental neurotoxicity induced by glutaraldehyde in neuron/astrocyte co-cultured cells and zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113891. [PMID: 35868176 DOI: 10.1016/j.ecoenv.2022.113891] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
The genotoxicity, development toxicity, carcinogenicity, and acute or chronic toxic effects of glutaraldehyde (GA), particularly during occupational exposure through its use as a fixative, disinfectant, and preservative, are well-documented but its effects on neurotoxicity have not been investigated. We performed in vitro and in vivo studies to examine the developmental neurotoxicity (DNT) of GA. Neurite outgrowth was examined in an in vitro co-culture model consisting of SH-SY5Y human neuroblastoma cells and human astrocytes. Cell Counting Kit-8, lactate dehydrogenase assay, and high-content screening revealed that GA significantly inhibited neurite outgrowth at non-cytotoxic concentration. Further studies showed that GA upregulated the mRNA expression of the astrocyte markers GFAP and S100β and downregulated the expression of the neurodevelopmental genes Nestin, βIII-tubulin, GAP43, and MAP2. Furthermore, in vivo zebrafish embryo toxicity tests explored the effects of GA on neural morphogenesis. GA adversely affected the early development of zebrafish embryos, resulting in decreased survival, irregular hatching, and reduced heart rate in a time- and concentration-dependent manner. Furthermore, the width of the brain and spinal cord was reduced, and the myelination of Schwann cells and oligodendrocytes was decreased by GA in transgenic zebrafish lines. These data suggest that GAs have potential DNT in vitro and in vivo, highlighting the need for caution regarding the neurotoxicity of GA.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Donggon Yoo
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Seungmin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Sangwoo Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
34
|
Mol P, Gopalakrishnan L, Chatterjee O, Mangalaparthi KK, Kumar M, Durgad SS, Nair B, Shankar SK, Mahadevan A, Prasad TSK. Proteomic Analysis of Adult Human Hippocampal Subfields Demonstrates Regional Heterogeneity in the Protein Expression. J Proteome Res 2022; 21:2293-2310. [PMID: 36039803 DOI: 10.1021/acs.jproteome.2c00143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background: Distinct hippocampal subfields are known to get affected during aging, psychiatric disorders, and various neurological and neurodegenerative conditions. To understand the biological processes associated with each subfield, it is important to understand its heterogeneity at the molecular level. To address this lacuna, we investigated the proteomic analysis of hippocampal subfields─the cornu ammonis sectors (CA1, CA2, CA3, CA4) and dentate gyrus (DG) from healthy adult human cohorts. Findings: Microdissection of hippocampal subfields from archived formalin-fixed paraffin-embedded tissue sections followed by TMT-based multiplexed proteomic analysis resulted in the identification of 5,593 proteins. Out of these, 890 proteins were found to be differentially abundant among the subfields. Further bioinformatics analysis suggested proteins related to gene splicing, transportation, myelination, structural activity, and learning processes to be differentially abundant in DG, CA4, CA3, CA2, and CA1, respectively. A subset of proteins was selected for immunohistochemistry-based validation in an independent set of hippocampal samples. Conclusions: We believe that our findings will effectively pave the way for further analysis of the hippocampal subdivisions and provide awareness of its subfield-specific association to various neurofunctional anomalies in the future. The current mass spectrometry data is deposited and publicly made available through ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD029697.
Collapse
Affiliation(s)
- Praseeda Mol
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore 560066,India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, India
| | - Lathika Gopalakrishnan
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore 560066,India.,Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore 560066,India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, India.,Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Kiran K Mangalaparthi
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore 560066,India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, India
| | - Manish Kumar
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore 560066,India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Shwetha S Durgad
- Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, India
| | - Susarla K Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | | |
Collapse
|
35
|
Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol 2022; 13:931473. [PMID: 35967420 PMCID: PMC9373849 DOI: 10.3389/fimmu.2022.931473] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a non-specific multi-ligand pattern recognition receptor capable of binding to a range of structurally diverse ligands, expressed on a variety of cell types, and performing different functions. The ligand-RAGE axis can trigger a range of signaling events that are associated with diabetes and its complications, neurological disorders, cancer, inflammation and other diseases. Since RAGE is involved in the pathophysiological processes of many diseases, targeting RAGE may be an effective strategy to block RAGE signaling.
Collapse
|
36
|
Chu C, Zhong R, Cai M, Li N, Lin W. Elevated Blood S100B Levels in Patients With Migraine: A Systematic Review and Meta-Analysis. Front Neurol 2022; 13:914051. [PMID: 35911929 PMCID: PMC9329586 DOI: 10.3389/fneur.2022.914051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In recent years, a growing number of researches indicate that S100B may act in migraine, but the relationship between S100B and migraine remains controversial. Therefore, the current study aimed to perform a meta-analysis to quantitatively summarize S100B levels in migraine patients. Methods We used Stata 12.0 software to summarize eligible studies from PubMed, EMBASE, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI), and Wanfang databases. We applied standardized mean differences (SMDs) with 95% confidence intervals (95%CIs) to appraise the association between S100B and migraine. Results The combined results of nine case-control studies indicated that compared with healthy controls, overall migraine patients had significantly increased S100B levels in peripheral blood (SMD = 0.688, 95%CI: 0.341–1.036, P < 0.001). The S100B levels in migraineurs during ictal periods (SMD =1.123, 95%CI: 0.409–1.836, P = 0.002) and interictal periods (SMD = 0.487, 95%CI: 0313–0.661, P < 0.001), aura (SMD = 0.999, 95%CI: 0.598–1.400, P < 0.001) and without aura (SMD = 0.534, 95%CI: 0.286–0.783, P < 0.001) were significantly higher than those in the controls. The subgroup analyses by age, country, migraine assessment, and assay method of S100B also illustrated a statistically obvious association between S100B levels and migraine, indicating that age may be the most important source of heterogeneity. Sensitivity analysis showed that no individual study has a significant influence on the overall association between S100B and migraine. Conclusion This meta-analysis demonstrates that the level of S100B in peripheral blood of patients with migraine was significantly increased. Migraine may be associated with pathological reactions involving S100B, which is instrumental for the clinical diagnosis of migraine and therapy that considers S100B as a potential target.
Collapse
|
37
|
Rocha GS, Freire MAM, Paiva KM, Oliveira RF, Norrara B, Morais PLAG, Oliveira LC, Engelberth RCGJ, Cavalcante JS, Cavalcanti JRLP. Effect of senescence on the tyrosine hydroxylase and S100B immunoreactivity in the nigrostriatal pathway of the rat. J Chem Neuroanat 2022; 124:102136. [PMID: 35809809 DOI: 10.1016/j.jchemneu.2022.102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Senescence is a natural and progressive physiological event that leads to a series of morphophysiological alterations in the organism. The brain is the most vulnerable organ to both structural and functional changes during this process. Dopamine is a key neurotransmitter for the proper functioning of the brain, directly involved in circuitries related with emotions, learning, motivation and reward. One of the main dopamine- producing nuclei is the substantia nigra pars compacta (SNpc), which establish connections with the striatum forming the so-called nigrostriatal pathway. S100B is a calcium binding protein mainly expressed by astrocytes, involved in both intracellular and extracellular processes, and whose expression is increased following injury in the nervous tissue, being a useful marker in altered status of central nervous system. The present study aimed to analyze the impact of senescence on the cells immunoreactive for tyrosine hydroxylase (TH) and S100B along the nigrostriatal pathway of the rat. Our results show an decreased expression of S100B+ cells in SNpc. In addition, there was a significant decrease in TH immunoreactivity in both projection fibers and TH+ cell bodies. In the striatum, a decrease in TH immunoreactivity was also observed, as well as an enlargement of the white matter bundles. Our findings point out that senescence is related to the anatomical and neurochemical changes observed throughout the nigrostriatal pathway.
Collapse
Affiliation(s)
- Gabriel S Rocha
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Marco Aurelio M Freire
- Graduate Program in Health and Society, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Karina M Paiva
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Rodrigo F Oliveira
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Bianca Norrara
- Laboratory of Experimental Neurology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Paulo Leonardo A G Morais
- Laboratory of Experimental Neurology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | - Lucidio C Oliveira
- Laboratory of Experimental Neurology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil
| | | | | | - José Rodolfo L P Cavalcanti
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil; Graduate Program in Health and Society, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil; Laboratory of Experimental Neurology, University of the State of Rio Grande do Norte (UERN), Mossoró, RN, Brazil.
| |
Collapse
|
38
|
Wong YY, Wu CY, Yu D, Kim E, Wong M, Elez R, Zebarth J, Ouk M, Tan J, Liao J, Haydarian E, Li S, Fang Y, Li P, Pakosh M, Tartaglia MC, Masellis M, Swardfager W. Biofluid markers of blood-brain barrier disruption and neurodegeneration in Lewy body spectrum diseases: A systematic review and meta-analysis. Parkinsonism Relat Disord 2022; 101:119-128. [PMID: 35760718 DOI: 10.1016/j.parkreldis.2022.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mixed evidence supports blood-brain barrier (BBB) dysfunction in Lewy body spectrum diseases. METHODS We compare biofluid markers in people with idiopathic Parkinson's disease (PD) and people with PD dementia (PDD) and/or dementia with Lewy bodies (DLB), compared with healthy controls (HC). Seven databases were searched up to May 10, 2021. Outcomes included cerebrospinal fluid to blood albumin ratio (Qalb), and concentrations of 7 blood protein markers that also reflect BBB disruption and/or neurodegenerative co-pathology. We further explore differences between PD patients with and without evidence of dementia. Random-effects models were used to obtain standardized mean differences (SMD) with 95% confidence interval. RESULTS Of 13,949 unique records, 51 studies were meta-analyzed. Compared to HC, Qalb was higher in PD (NPD/NHC = 224/563; SMD = 0.960 [0.227-1.694], p = 0.010; I2 = 92.2%) and in PDD/DLB (NPDD/DLB/NHC = 265/670; SMD = 1.126 [0.358-1.893], p < 0.001; I2 = 78.2%). Blood neurofilament light chain (NfL) was higher in PD (NPD/NHC = 1848/1130; SMD = 0.747 [0.442-1.052], p < 0.001; I2 = 91.9%) and PDD/DLB (NPDD/DLB/NHC = 183/469; SMD = 1.051 [0.678-1.423], p = 0.004; I2 = 92.7%) than in HC. p-tau 181 (NPD/NHC = 276/164; SMD = 0.698 [0.149-1.247], p = 0.013; I2 = 82.7%) was also higher in PD compared to HC. In exploratory analyses, blood NfL was higher in PD without dementia (NPDND/NHC = 1005/740; SMD = 0.252 [0.042-0.462], p = 0.018; I2 = 71.8%) and higher in PDD (NPDD/NHC = 100/111; SMD = 0.780 [0.347-1.214], p < 0.001; I2 = 46.7%) compared to HC. Qalb (NPDD/NPDND = 63/191; SMD = 0.482 [0.189-0.774], p = 0.010; I2<0.001%) and NfL (NPDD/NPDND = 100/223; SMD = 0.595 [0.346-0.844], p < 0.001; I2 = 3.4%) were higher in PDD than in PD without dementia. CONCLUSIONS Biofluid markers suggest BBB disruption and neurodegenerative co-pathology involvement in common Lewy body diseases. Greater evidence of BBB breakdown was seen in Lewy body disease with cognitive impairment.
Collapse
Affiliation(s)
- Yuen Yan Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Di Yu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Esther Kim
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Renata Elez
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Zebarth
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Ouk
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jocelyn Tan
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jiamin Liao
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Eileen Haydarian
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Siming Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Yaolu Fang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Peihao Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Maureen Pakosh
- Library & Information Services, UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Mario Masellis
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada.
| |
Collapse
|
39
|
Li Y, Peng Y, Shen Y, Zhang Y, Liu L, Yang X. Dietary polyphenols: regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit Rev Food Sci Nutr 2022; 63:9816-9842. [PMID: 35587161 DOI: 10.1080/10408398.2022.2076064] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Advanced glycation end products (AGEs) are formed in non-enzymatic reaction, oxidation, rearrangement and cross-linking between the active carbonyl groups of reducing sugars and the free amines of amino acids. The Maillard reaction is related to sensory characteristics in thermal processed food, while AGEs are formed in food matrix in this process. AGEs are a key link between carbonyl stress and neurodegenerative disease. AGEs can interact with receptors for AGEs (RAGE), causing oxidative stress, inflammation response and signal pathways activation related to neurodegenerative diseases. Neurodegenerative diseases are closely related to gut microbiota imbalance and intestinal inflammation. Polyphenols with multiple hydroxyl groups showed a powerful ability to scavenge ROS and capture α-dicarbonyl species, which led to the formation of mono- and di- adducts, thereby inhibiting AGEs formation. Neurodegenerative diseases can be effectively prevented by inhibiting AGEs production, and interaction with RAGEs, or regulating the microbiota-gut-brain axis. These strategies include polyphenols multifunctional effects on AGEs inhibition, RAGE-ligand interactions blocking, and regulating the abundance and diversity of gut microbiota, and intestinal inflammation alleviation to delay or prevent neurodegenerative diseases progress. It is a wise and promising strategy to supplement dietary polyphenols for preventing neurodegenerative diseases via AGEs-RAGE axis and microbiota-gut-brain axis regulation.
Collapse
Affiliation(s)
- Yueqin Li
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Yao Peng
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, Guangdong, PR China
| | - Yingbin Shen
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, PR China
| | - Yunzhen Zhang
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Lianliang Liu
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Xinquan Yang
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, Guangdong, PR China
| |
Collapse
|
40
|
Smajić S, Prada-Medina CA, Landoulsi Z, Ghelfi J, Delcambre S, Dietrich C, Jarazo J, Henck J, Balachandran S, Pachchek S, Morris CM, Antony P, Timmermann B, Sauer S, Pereira SL, Schwamborn JC, May P, Grünewald A, Spielmann M. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain 2022; 145:964-978. [PMID: 34919646 PMCID: PMC9050543 DOI: 10.1093/brain/awab446] [Citation(s) in RCA: 195] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/21/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022] Open
Abstract
Idiopathic Parkinson's disease is characterized by a progressive loss of dopaminergic neurons, but the exact disease aetiology remains largely unknown. To date, Parkinson's disease research has mainly focused on nigral dopaminergic neurons, although recent studies suggest disease-related changes also in non-neuronal cells and in midbrain regions beyond the substantia nigra. While there is some evidence for glial involvement in Parkinson's disease, the molecular mechanisms remain poorly understood. The aim of this study was to characterize the contribution of all cell types of the midbrain to Parkinson's disease pathology by single-nuclei RNA sequencing and to assess the cell type-specific risk for Parkinson's disease using the latest genome-wide association study. We profiled >41 000 single-nuclei transcriptomes of post-mortem midbrain from six idiopathic Parkinson's disease patients and five age-/sex-matched controls. To validate our findings in a spatial context, we utilized immunolabelling of the same tissues. Moreover, we analysed Parkinson's disease-associated risk enrichment in genes with cell type-specific expression patterns. We discovered a neuronal cell cluster characterized by CADPS2 overexpression and low TH levels, which was exclusively present in idiopathic Parkinson's disease midbrains. Validation analyses in laser-microdissected neurons suggest that this cluster represents dysfunctional dopaminergic neurons. With regard to glial cells, we observed an increase in nigral microglia in Parkinson's disease patients. Moreover, nigral idiopathic Parkinson's disease microglia were more amoeboid, indicating an activated state. We also discovered a reduction in idiopathic Parkinson's disease oligodendrocyte numbers with the remaining cells being characterized by a stress-induced upregulation of S100B. Parkinson's disease risk variants were associated with glia- and neuron-specific gene expression patterns in idiopathic Parkinson's disease cases. Furthermore, astrocytes and microglia presented idiopathic Parkinson's disease-specific cell proliferation and dysregulation of genes related to unfolded protein response and cytokine signalling. While reactive patient astrocytes showed CD44 overexpression, idiopathic Parkinson's disease microglia revealed a pro-inflammatory trajectory characterized by elevated levels of IL1B, GPNMB and HSP90AA1. Taken together, we generated the first single-nuclei RNA sequencing dataset from the idiopathic Parkinson's disease midbrain, which highlights a disease-specific neuronal cell cluster as well as 'pan-glial' activation as a central mechanism in the pathology of the movement disorder. This finding warrants further research into inflammatory signalling and immunomodulatory treatments in Parkinson's disease.
Collapse
Affiliation(s)
- Semra Smajić
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | | | - Zied Landoulsi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jenny Ghelfi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Carola Dietrich
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jana Henck
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | | | - Sinthuja Pachchek
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Christopher M. Morris
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, NE1 7RU Newcastle upon Tyne, UK
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | - Sascha Sauer
- Max-Delbrück-Centrum für Molekulare Medizin, Genomics Group, D-13125 Berlin, Germany
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, L-4362 Esch-sur-Alzette, Luxembourg
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, D-23562 Lübeck, Germany
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
- Institute of Human Genetics, Kiel University, D-42118 Kiel, Germany
- Institute of Human Genetics, University of Lübeck, D-23562 Lübeck, Germany
| |
Collapse
|
41
|
Weiss F, Labrador-Garrido A, Dzamko N, Halliday G. Immune responses in the Parkrtdinson's disease brain. Neurobiol Dis 2022; 168:105700. [DOI: 10.1016/j.nbd.2022.105700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
|
42
|
Kinscherf NA, Pehar M. Role and Therapeutic Potential of RAGE Signaling in Neurodegeneration. Curr Drug Targets 2022; 23:1191-1209. [PMID: 35702767 PMCID: PMC9589927 DOI: 10.2174/1389450123666220610171005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023]
Abstract
Activation of the receptor for advanced glycation end products (RAGE) has been shown to play an active role in the development of multiple neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Although originally identified as a receptor for advanced glycation end products, RAGE is a pattern recognition receptor able to bind multiple ligands. The final outcome of RAGE signaling is defined in a context and cell type specific manner and can exert both neurotoxic and neuroprotective functions. Contributing to the complexity of the RAGE signaling network, different RAGE isoforms with distinctive signaling capabilities have been described. Moreover, multiple RAGE ligands bind other receptors and RAGE antagonism can significantly affect their signaling. Here, we discuss the outcome of celltype specific RAGE signaling in neurodegenerative pathologies. In addition, we will review the different approaches that have been developed to target RAGE signaling and their therapeutic potential. A clear understanding of the outcome of RAGE signaling in a cell type- and disease-specific manner would contribute to advancing the development of new therapies targeting RAGE. The ability to counteract RAGE neurotoxic signaling while preserving its neuroprotective effects would be critical for the success of novel therapies targeting RAGE signaling.
Collapse
Affiliation(s)
- Noah Alexander Kinscherf
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mariana Pehar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, USA
| |
Collapse
|
43
|
Costas C, Faro LR. Do Naturally Occurring Antioxidants Protect Against Neurodegeneration of the Dopaminergic System? A Systematic Revision in Animal Models of Parkinson's Disease. Curr Neuropharmacol 2022; 20:432-459. [PMID: 33882808 PMCID: PMC9413795 DOI: 10.2174/1570159x19666210421092725] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by a significant decrease in dopamine levels, caused by progressive degeneration of the dopaminergic neurons in the nigrostriatal pathway. Multiple mechanisms have been implicated in its pathogenesis, including oxidative stress, neuroinflammation, protein aggregation, mitochondrial dysfunction, insufficient support for neurotrophic factors and cell apoptosis. The absence of treatments capable of slowing or stopping the progression of PD has increased the interest in the natural antioxidant substances present in the diet, since they have multiple beneficial properties and it is possible that they can influence the mechanisms responsible for the dysfunction and death of dopaminergic neurons. Thus, the purpose of this systematic review is to analyze the results obtained in a set of studies carried out in the last years, which describe the neuroprotective, antioxidant and regenerative functions of some naturally occurring antioxidants in experimental models of PD. The results show that the exogenous no enzymatic antioxidants can significantly modify the biochemical and behavioral mechanisms that contribute to the pathophysiology of Parkinsonism in experimental animals. Therefore, it is possible that they may contribute to effective neuroprotection by providing a significant improvement in neuropathological markers. In conclusion, the results of this review suggest that exogenous antioxidants can be promising therapeutic candidates for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Carmen Costas
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| | - Lilian R.F. Faro
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| |
Collapse
|
44
|
Zimmermann M, Brockmann K. Blood and Cerebrospinal Fluid Biomarkers of Inflammation in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S183-S200. [PMID: 35661021 PMCID: PMC9535573 DOI: 10.3233/jpd-223277] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 02/07/2023]
Abstract
Given the clear role of inflammation in the pathogenesis of Parkinson's disease (PD) and its impact on incidence and phenotypical characteristics, this review provides an overview with focus on inflammatory biofluid markers in blood and cerebrospinal fluid (CSF) in PD patient cohorts. In preparation for clinical trials targeting the immune system, we specifically address the following questions: 1) What evidence do we have for pro-inflammatory profiles in blood and in CSF of sporadic and genetic PD patients? 2) Is there a role of anti-inflammatory mediators in blood/CSF? 3) Do inflammatory profiles in blood reflect those in CSF indicative of a cross-talk between periphery and brain? 4) Do blood/CSF inflammatory profiles change over the disease course as assessed in repeatedly taken biosamples? 5) Are blood/CSF inflammatory profiles associated with phenotypical trajectories in PD? 6) Are blood/CSF inflammatory profiles associated with CSF levels of neurodegenerative/PD-specific biomarkers? Knowledge on these questions will inform future strategies for patient stratification and cohort enrichment as well as suitable outcome measures for clinical trials.
Collapse
Affiliation(s)
- Milan Zimmermann
- Center of Neurology, Department of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Kathrin Brockmann
- Center of Neurology, Department of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
45
|
Augusto-Oliveira M, Arrifano GDP, Lopes-Araújo A, Santos-Sacramento L, Lima RR, Lamers ML, Le Blond J, Crespo-Lopez ME. Salivary biomarkers and neuropsychological outcomes: A non-invasive approach to investigate pollutants-associated neurotoxicity and its effects on cognition in vulnerable populations. ENVIRONMENTAL RESEARCH 2021; 200:111432. [PMID: 34062204 DOI: 10.1016/j.envres.2021.111432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 06/12/2023]
Abstract
The occurrence of neurotoxicity caused by xenobiotics such as pesticides (dichlorodiphenyltrichloroethane, organophosphates, pyrethroids, etc.) or metals (mercury, lead, aluminum, arsenic, etc.) is a growing concern around the world, particularly in vulnerable populations with difficulties on both detection and symptoms treatment, due to low economic status, remote access, poor infrastructure, and low educational level, among others features. Despite the numerous molecular markers and questionnaires/clinical evaluations, studying neurotoxicity and its effects on cognition in these populations faces problems with samples collection and processing, and information accuracy. Assessing cognitive changes caused by neurotoxicity, especially those that are subtle in the initial stages, is fundamentally challenging. Finding accurate, non-invasive, and low-cost strategies to detect the first signals of brain injury has the potential to support an accelerated development of the research with these populations. Saliva emerges as an ideal pool of biomarkers (with interleukins and neural damage-related proteins, among others) and potential alternative diagnostic fluid to molecularly investigate neurotoxicity. As a source of numerous neurological biomarkers, saliva has several advantages compared to blood, such as easier storage, requires less manipulation, and the procedure is cheaper, safer and well accepted by patients compared with drawing blood. Regarding cognitive dysfunction, neuropsychological batteries represent, with their friendly interface, a feasible and accurate method to evaluate the eventual cognitive deficits associated with neurotoxicity in people from diverse cultural and educational backgrounds. The association of these two tools, saliva and neuropsychological batteries, to cover the molecular and cognitive aspects of neurotoxicity in vulnerable populations, could potentially increase the prevalence of early intervention and successful treatment.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| | - Letícia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| | - Rafael Rodrigues Lima
- Laboratório de Biologia Estrutural e Funcional, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| | - Marcelo Lazzaron Lamers
- Department of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil.
| | | | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, 66075-110, Brazil.
| |
Collapse
|
46
|
Navarro E, Udine E, Lopes KDP, Parks M, Riboldi G, Schilder BM, Humphrey J, Snijders GJL, Vialle RA, Zhuang M, Sikder T, Argyrou C, Allan A, Chao MJ, Farrell K, Henderson B, Simon S, Raymond D, Elango S, Ortega RA, Shanker V, Swan M, Zhu CW, Ramdhani R, Walker RH, Tse W, Sano M, Pereira AC, Ahfeldt T, Goate AM, Bressman S, Crary JF, de Witte L, Frucht S, Saunders-Pullman R, Raj T. Dysregulation of mitochondrial and proteolysosomal genes in Parkinson's disease myeloid cells. NATURE AGING 2021; 1:850-863. [PMID: 35005630 PMCID: PMC8728893 DOI: 10.1038/s43587-021-00110-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022]
Abstract
An increasing number of identified Parkinson's disease (PD) risk loci contain genes highly expressed in innate immune cells, yet their role in pathology is not understood. We hypothesize that PD susceptibility genes modulate disease risk by influencing gene expression within immune cells. To address this, we have generated transcriptomic profiles of monocytes from 230 individuals with sporadic PD and healthy subjects. We observed a dysregulation of mitochondrial and proteasomal pathways. We also generated transcriptomic profiles of primary microglia from brains of 55 subjects and observed discordant transcriptomic signatures of mitochondrial genes in PD monocytes and microglia. We further identified 17 PD susceptibility genes whose expression, relative to each risk allele, is altered in monocytes. These findings reveal widespread transcriptomic alterations in PD monocytes, with some being distinct from microglia, and facilitate efforts to understand the roles of myeloid cells in PD as well as the development of biomarkers.
Collapse
Affiliation(s)
- Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evan Udine
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulietta Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
- Universita degli Studi di Milano, Molecular and Translational Medicine, Milan, Italy
| | - Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gijsje J L Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, NY, Bronx, USA
| | - Ricardo A Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maojuan Zhuang
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tamjeed Sikder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charalambos Argyrou
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael J Chao
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kurt Farrell
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brooklyn Henderson
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Sarah Simon
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deborah Raymond
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonya Elango
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto A Ortega
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vicki Shanker
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Swan
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn W Zhu
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Centers, James J. Peters VA Medical Center, New York, NY, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ritesh Ramdhani
- Department of Neurology, Zucker School of Medicine at Hofstra Northwell, New York, NY, USA
| | - Ruth H Walker
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Winona Tse
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Sano
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Centers, James J. Peters VA Medical Center, New York, NY, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana C Pereira
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tim Ahfeldt
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Bressman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Crary
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lotje de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, NY, Bronx, USA
| | - Steven Frucht
- The Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Rachel Saunders-Pullman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
47
|
Michetti F, Di Sante G, Clementi ME, Sampaolese B, Casalbore P, Volonté C, Romano Spica V, Parnigotto PP, Di Liddo R, Amadio S, Ria F. Growing role of S100B protein as a putative therapeutic target for neurological- and nonneurological-disorders. Neurosci Biobehav Rev 2021; 127:446-458. [PMID: 33971224 DOI: 10.1016/j.neubiorev.2021.04.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023]
Abstract
S100B is a calcium-binding protein mainly expressed by astrocytes, but also localized in other definite neural and extra-neural cell types. While its presence in biological fluids is widely recognized as a reliable biomarker of active injury, growing evidence now indicates that high levels of S100B are suggestive of pathogenic processes in different neural, but also extra-neural, disorders. Indeed, modulation of S100B levels correlates with the occurrence of clinical and/or toxic parameters in experimental models of diseases such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, muscular dystrophy, multiple sclerosis, acute neural injury, inflammatory bowel disease, uveal and retinal disorders, obesity, diabetes and cancer, thus directly linking the levels of S100B to pathogenic mechanisms. In general, deletion/inactivation of the protein causes the improvement of the disease, whereas its over-expression/administration induces a worse clinical presentation. This scenario reasonably proposes S100B as a common therapeutic target for several different disorders, also offering new clues to individuate possible unexpected connections among these diseases.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| | - Gabriele Di Sante
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168 Rome, Italy.
| | - Maria Elisabetta Clementi
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Beatrice Sampaolese
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Patrizia Casalbore
- Institute for Systems Analysis and Computer Science, IASI-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Cinzia Volonté
- Institute for Systems Analysis and Computer Science, IASI-CNR, Largo Francesco Vito 1, 00168 Rome, Italy; Cellular Neurobiology Unit, Preclinical Neuroscience, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Vincenzo Romano Spica
- Department of Movement, Human and Health Sciences, Laboratory of Epidemiology and Biotechnologies, University of Rome "Foro Italico", Piazza Lauro De Bosis 6, 00135, Rome, Italy.
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, Padua, Italy.
| | - Rosa Di Liddo
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, Padua, Italy; Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy.
| | - Susanna Amadio
- Cellular Neurobiology Unit, Preclinical Neuroscience, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168 Rome, Italy.
| |
Collapse
|
48
|
Gönen M, Özdoğan S, Balgetir F, Demir CF, Aytaç E, Müngen B. S100B and neuron-specific enolase levels in episodic and chronic migraine. Acta Neurol Scand 2021; 143:298-302. [PMID: 33089498 DOI: 10.1111/ane.13365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/19/2020] [Accepted: 10/16/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES In recent years, radiological and biochemical data have emerged regarding the development of cellular damage in the brain of patients with migraine, calling into question what has traditionally been accepted as a benign disorder. In order to investigate whether cellular damage develops in the brain of episodic migraine patient, serum levels of neuron-specific enolase (NSE) and S100B have been evaluated in recent studies. However, contradictory results were obtained in these studies. Moreover, there is no study on NSE and S100B in cases of chronic migraine. METHODS Patients with episodic migraine with or without aura and chronic migraine were included. In addition, 27 healthy volunteers were included as a control group. Control group was selected from healthy volunteers of the same age and sex. We investigated serum NSE and S100B levels during the interictal period in 26 patients with episodic migraine and 27 patients with chronic migraine. RESULTS The serum NSE and S100B levels were significantly higher in both patients with episodic and chronic migraine than controls. Although there were no significant differences in the serum NSE and S100B levels between the two patients' groups, these markers were found to be higher in cases of chronic migraine. CONCLUSION These results suggest that there is both neuronal and glial involvement in the two migraine groups. Elevations in these markers in cases of episodic migraine suggest that cellular damage not only results from headache episodes, but that there may be also an ongoing pathological process during the interictal period.
Collapse
Affiliation(s)
- Murat Gönen
- Department of Neurology, Faculty of Medicine Firat University Elazig Turkey
| | - Sevim Özdoğan
- Department of Neurology Kayseri City Hospital Kayseri Turkey
| | - Ferhat Balgetir
- Department of Neurology, Faculty of Medicine Firat University Elazig Turkey
| | - Caner Feyzi Demir
- Department of Neurology, Faculty of Medicine Firat University Elazig Turkey
| | - Emrah Aytaç
- Department of Neurology, Faculty of Medicine Firat University Elazig Turkey
| | - Bülent Müngen
- Department of Neurology, Faculty of Medicine Firat University Elazig Turkey
| |
Collapse
|
49
|
Rungratanawanich W, Qu Y, Wang X, Essa MM, Song BJ. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury. Exp Mol Med 2021; 53:168-188. [PMID: 33568752 PMCID: PMC8080618 DOI: 10.1038/s12276-021-00561-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 01/30/2023] Open
Abstract
Advanced glycation end products (AGEs) are potentially harmful and heterogeneous molecules derived from nonenzymatic glycation. The pathological implications of AGEs are ascribed to their ability to promote oxidative stress, inflammation, and apoptosis. Recent studies in basic and translational research have revealed the contributing roles of AGEs in the development and progression of various aging-related pathological conditions, such as diabetes, cardiovascular complications, gut microbiome-associated illnesses, liver or neurodegenerative diseases, and cancer. Excessive chronic and/or acute binge consumption of alcohol (ethanol), a widely consumed addictive substance, is known to cause more than 200 diseases, including alcohol use disorder (addiction), alcoholic liver disease, and brain damage. However, despite the considerable amount of research in this area, the underlying molecular mechanisms by which alcohol abuse causes cellular toxicity and organ damage remain to be further characterized. In this review, we first briefly describe the properties of AGEs: their formation, accumulation, and receptor interactions. We then focus on the causative functions of AGEs that impact various aging-related diseases. We also highlight the biological connection of AGE-alcohol-adduct formations to alcohol-mediated tissue injury. Finally, we describe the potential translational research opportunities for treatment of various AGE- and/or alcohol-related adduct-associated disorders according to the mechanistic insights presented.
Collapse
Affiliation(s)
- Wiramon Rungratanawanich
- grid.420085.b0000 0004 0481 4802Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Ying Qu
- grid.420085.b0000 0004 0481 4802Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892 USA
| | - Xin Wang
- Neuroapoptosis Drug Discovery Laboratory, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115 USA
| | - Musthafa Mohamed Essa
- grid.412846.d0000 0001 0726 9430Department of Food Science and Nutrition, Aging and Dementia Research Group, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khoud, Muscat, Oman ,grid.412846.d0000 0001 0726 9430Aging and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| | - Byoung-Joon Song
- grid.420085.b0000 0004 0481 4802Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892 USA
| |
Collapse
|
50
|
Angelopoulou E, Paudel YN, Piperi C. Emerging role of S100B protein implication in Parkinson's disease pathogenesis. Cell Mol Life Sci 2021; 78:1445-1453. [PMID: 33052436 PMCID: PMC11073186 DOI: 10.1007/s00018-020-03673-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/10/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
The exact etiology of Parkinson's disease (PD) remains obscure, lacking effective diagnostic and prognostic biomarkers. In search of novel molecular factors that may contribute to PD pathogenesis, emerging evidence highlights the multifunctional role of the calcium-binding protein S100B that is widely expressed in the brain and predominantly in astrocytes. Preclinical evidence points towards the possible time-specific contributing role of S100B in the pathogenesis of neurodegenerative disorders including PD, mainly by regulating neuroinflammation and dopamine metabolism. Although existing clinical evidence presents some contradictions, estimation of S100B in the serum and cerebrospinal fluid seems to hold a great promise as a potential PD biomarker, particularly regarding the severity of motor and non-motor PD symptoms. Furthermore, given the recent development of S100B inhibitors that are able to cross the blood brain barrier, novel opportunities are arising in the research field of PD therapeutics. In this review, we provide an update on recent advances in the implication of S100B protein in the pathogenesis of PD and discuss relevant studies investigating the biomarker potential of S100B in PD, aiming to shed more light on clinical targeting approaches related to this incurable disorder.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527, Athens, Greece.
| |
Collapse
|