1
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. NPJ Parkinsons Dis 2025; 11:77. [PMID: 40240380 PMCID: PMC12003903 DOI: 10.1038/s41531-025-00933-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Many drug targets in ongoing Parkinson's disease (PD) clinical trials have strong genetic links. While genome-wide association studies (GWAS) nominate regions associated with disease, pinpointing causal genes is challenging. Our aim was to prioritize additional druggable genes underlying PD GWAS signals. The polygenic priority score (PoPS) integrates genome-wide information from MAGMA gene-level associations and over 57,000 gene-level features. We applied PoPS to East Asian and European PD GWAS data and prioritized genes based on PoPS, distance to the GWAS signal, and non-synonymous credible set variants. We prioritized 46 genes, including well-established PD genes (SNCA, LRRK2, GBA1, TMEM175, VPS13C), genes with strong literature evidence supporting a mechanistic link to PD (RIT2, BAG3, SCARB2, FYN, DYRK1A, NOD2, CTSB, SV2C, ITPKB), and genes relatively unexplored in PD. Many hold potential for drug repurposing or development. We prioritized high-confidence genes with strong links to PD pathogenesis that may represent our next-best candidates for developing disease-modifying therapeutics.
Collapse
Affiliation(s)
- Lara M Lange
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- Centre de recherche du Centre Hospitalier Universitaire de Québec, Axe Neurosciences, Département de Psychiatrie et Neurosciences, Laval University, Québec, QC, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany.
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany.
| |
Collapse
|
2
|
Zhang S, Zhang C, Zhang Y, Feng Y. Unraveling the role of neuregulin-mediated astrocytes-OPCs axis in the pathogenesis of age-related macular degeneration and Parkinson's disease. Sci Rep 2025; 15:7352. [PMID: 40025106 PMCID: PMC11873146 DOI: 10.1038/s41598-025-92103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Age-related macular degeneration (AMD) and Parkinson's disease (PD) are prevalent and debilitating conditions that lead to irreversible blindness and dyskinesia, respectively. Emerging evidences imply that retinal abnormalities may serve as early indicators for monitoring PD. This study endeavors to explore the complex interactions and focus on their shared molecular and pathological mechanisms. We employed a comprehensive approach by integrating single-cell RNA sequencing (scRNA-seq) datasets, obtained from dry AMD retinas and PD brain tissues, along with Weighted Gene Co-expression Network Analysis (WGCNA)-related computational analysis. Gene Set Enrichment Analysis (GSEA) was conducted to analyze PD-related genes within retinal ganglion cells in dry AMD. Cell-cell chat was utilized to predict intercellular communication and signaling pathways. Module eigengenes (MEs) were calculated to identify specific gene modules. Dysregulation of PALLD, FYN and ZMZ1 may lead to cell structural abnormalities, impaired mitochondrial functions, and increased susceptibility to neuroinflammation, contributing to the AMD and PD progression. Additionally, this study highlighted an astrocyte-oligodendrocyte precursor cell (OPCs) signaling axis mediated by Neuregulin (NRG), which is hypothesized to influence neuroinflammatory processes characteristic of dry AMD and PD pathogenesis. Notably, ME-salmon module associated with gene dysregulation exhibited a strong positive correlation with the ME-blue module, linked to neurodegenerative impairment, and the ME-yellow module, related to mitochondrial dysfunction. The comprehensive investigation on astrocytes-OPCs signaling axis, and the NRG signaling pathway advances our understanding of the intricate biological processes underpinning AMD and PD. This research underscores the critical importance about exploring glial-related cell interactions, providing valuable insights into potential novel therapeutic approaches for these complex diseases.
Collapse
Affiliation(s)
- Shuyan Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yinjian Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Zamanian MY, Kamran Z, Tavakoli MR, Oghenemaro EF, Abohassan M, Kubaev A, Nathiya D, Kaur P, Zwamel AH, Abdulamer RS. The Role of ΔFosB in the Pathogenesis of Levodopa-Induced Dyskinesia: Mechanisms and Therapeutic Strategies. Mol Neurobiol 2025:10.1007/s12035-025-04720-z. [PMID: 39890697 DOI: 10.1007/s12035-025-04720-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] [Received: 09/13/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Levodopa-induced dyskinesia (LID) represents a significant complication associated with the long-term administration of levodopa (L-DOPA) for the treatment of Parkinson's disease (PD). This review examines the critical role of ΔFosB, a transcription factor, in the pathogenesis of LID and explores potential therapeutic interventions. ΔFosB accumulates within the striatum in response to chronic dopaminergic stimulation, thereby driving maladaptive changes that culminate in dyskinesia. Its persistent expression modifies gene transcription, influencing neuronal plasticity and contributing to the sustained presence of dyskinetic movements. This study explains how ΔFosB functions at the molecular level, focusing on its connections with dopamine D1 receptors, the cAMP/PKA signaling pathway, and its regulatory effects on downstream targets such as DARPP-32 and GluA1 AMPA receptor subunits. Additionally, it examines how neuronal nitric oxide synthase (nNOS) affects ΔFosB levels and the development of LID. This review also considers the interactions between ΔFosB and other signaling pathways, such as ERK and mTOR, in the context of LID and striatal plasticity. Emerging therapeutic strategies targeting ΔFosB and its associated pathways include pharmacological interventions like ranitidine, 5-hydroxytryptophan, and carnosic acid. Furthermore, this study addresses the role of JunD, another component of the AP-1 transcription factor complex, in the pathogenesis of LID. Understanding the molecular mechanisms by which ΔFosB contributes to LID offers promising avenues for developing novel treatments that could mitigate dyskinesia and improve the quality of life for PD patients undergoing long-term L-DOPA therapy.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Zahra Kamran
- Department of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marziye Ranjbar Tavakoli
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology & Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Resan Shakir Abdulamer
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
4
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.13.24318996. [PMID: 39711693 PMCID: PMC11661345 DOI: 10.1101/2024.12.13.24318996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Recent advancements in Parkinson's disease (PD) drug development have been significantly driven by genetic research. Importantly, drugs supported by genetic evidence are more likely to be approved. While genome-wide association studies (GWAS) are a powerful tool to nominate genomic regions associated with certain traits or diseases, pinpointing the causal biologically relevant gene is often challenging. Our aim was to prioritize genes underlying PD GWAS signals. The polygenic priority score (PoPS) is a similarity-based gene prioritization method that integrates genome-wide information from MAGMA gene-level association tests and more than 57,000 gene-level features, including gene expression, biological pathways, and protein-protein interactions. We applied PoPS to data from the largest published PD GWAS in East Asian- and European-ancestries. We identified 120 independent associations with P < 5×10-8 and prioritized 46 PD genes across these loci based on their PoPS scores, distance to the GWAS signal, and presence of non-synonymous variants in the credible set. Alongside well-established PD genes (e.g., TMEM175 and VPS13C), some of which are targeted in ongoing clinical trials (i.e., SNCA, LRRK2, and GBA1), we prioritized genes with a plausible mechanistic link to PD pathogenesis (e.g., RIT2, BAG3, and SCARB2). Many of these genes hold potential for drug repurposing or novel therapeutic developments for PD (i.e., FYN, DYRK1A, NOD2, CTSB, SV2C, and ITPKB). Additionally, we prioritized potentially druggable genes that are relatively unexplored in PD (XPO1, PIK3CA, EP300, MAP4K4, CAMK2D, NCOR1, and WDR43). We prioritized a high-confidence list of genes with strong links to PD pathogenesis that may represent our next-best candidates for disease-modifying therapeutics. We hope our findings stimulate further investigations and preclinical work to facilitate PD drug development programs.
Collapse
Affiliation(s)
- Lara M. Lange
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- CHU de Québec Research Center, Axe Neurosciences, Laval University, Quebec City, Quebec, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
- Current address: Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany
| |
Collapse
|
5
|
Zhou J, Lu X, Wang H. The underlying molecular mechanisms of Fyn in neonatal hypoxic-ischaemic encephalopathy. Front Cell Neurosci 2024; 18:1476856. [PMID: 39664999 PMCID: PMC11631624 DOI: 10.3389/fncel.2024.1476856] [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: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Fyn is a cytoplasmic tyrosine kinase (TK) that is a nonreceptor and a member of the Src family of kinases (SFKs). It is involved in several transduction pathways in the central nervous system (CNS), such as oligodendrocyte development, myelination, axon guidance, and synaptic transmission. Owing to its wide range of activities in the molecular signaling pathways that underpin both neuropathologic and neurodevelopmental events, Fyn has remained of great interest for more than a century. Accumulating preclinical data have highlighted the potential role of Fyn in the pathophysiology of neonatal hypoxic-ischaemic encephalopathy (HIE). By mediating important signaling pathways, Fyn may control glutamate excitotoxicity, promote neuroinflammation and facilitate the death of neurons caused by oxidative stress. In this review, we address new evidence regarding the role of Fyn in the pathogenesis of this condition, with the aim of providing a reference for the development of new strategies to improve the prognosis of neonatal HIE. In addition, we also offer insights into additional Fyn-related molecular mechanisms involved in HIE pathology.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Reproductive Medicine Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiang Lu
- Department of Cardiology, The First People’s Hospital of Yuexi County, Yuexi, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Tan Y, Cheng C, Zheng C, Zeng W, Yang X, Xu Y, Zhang Z, Ma Z, Xu Y, Cao X. Activation of mGlu 2/3 receptors in the striatum alleviates L-DOPA-induced dyskinesia and inhibits abnormal postsynaptic molecular expression. Pharmacol Biochem Behav 2023; 231:173637. [PMID: 37714223 DOI: 10.1016/j.pbb.2023.173637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Group II metabotropic glutamate receptors (mGlu2/3 receptors) have been regarded as promising candidates for the treatment of L-DOPA-induced dyskinesia (LID); however, confirmation is still lacking. As the hub of the basal ganglia circuit, the striatum plays a critical role in action control. Supersensitive responsiveness of glutamatergic corticostriatal input may be the key mechanism for the development of LID. In this study, we first examined the potency of LY354740 (12 mg/kg, i.p.) in modulating glutamate and dopamine release in lesioned striatum of stable LID rats. Then, we injected LY354740 (20nmoL or 40nmoL in 4 μL of sterile 0.9 % saline) directly into the lesioned striatum to verify its ability to reduce or attenuate L-DOPA-induced abnormal involuntary movements. In experiment conducted in established LID rats, after continuous injection for 4 days, we found that LY354740 significantly reduced the expression of dyskinesia. In another experiment conducted in parkinsonism rat models, we found that LY354740 attenuated the development of LID with an inverted-U dose-response curve. The role of LY354740 in modulating striatal expressions of LID-related molecular changes was also assessed after these behavioral experiments. We found that LY354740 significantly inhibited abnormal expressions of p-Fyn/p-NMDA/p-ERK1/2/p-HistoneH3/ΔFosB, which is in line with its ability to alleviate abnormal involuntary movements in both LID expression and induction phase. Our study indicates that activation of striatal mGlu2/3 receptors can attenuate the development of dyskinesia in parkinsonism rats and provide some functional improvements in LID rats by inhibiting LID-related molecular changes.
Collapse
Affiliation(s)
- Yang Tan
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Chi Cheng
- Department of Neurology, Hanchuan People's Hospital, 432300, China
| | - Cong Zheng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200000, China
| | - Weiqi Zeng
- Department of Neurology, The First People's Hospital of Foshan, Foshan 528000, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yu Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Zhaoyuan Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
7
|
Guglietti B, Sivasankar S, Mustafa S, Corrigan F, Collins-Praino LE. Fyn Kinase Activity and Its Role in Neurodegenerative Disease Pathology: a Potential Universal Target? Mol Neurobiol 2021; 58:5986-6005. [PMID: 34432266 DOI: 10.1007/s12035-021-02518-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Fyn is a non-receptor tyrosine kinase belonging to the Src family of kinases (SFKs) which has been implicated in several integral functions throughout the central nervous system (CNS), including myelination and synaptic transmission. More recently, Fyn dysfunction has been associated with pathological processes observed in neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). Neurodegenerative diseases are amongst the leading cause of death and disability worldwide and, due to the ageing population, prevalence is predicted to rise in the coming years. Symptoms across neurodegenerative diseases are both debilitating and degenerative in nature and, concerningly, there are currently no disease-modifying therapies to prevent their progression. As such, it is important to identify potential new therapeutic targets. This review will outline the role of Fyn in normal/homeostatic processes, as well as degenerative/pathological mechanisms associated with neurodegenerative diseases, such as demyelination, pathological protein aggregation, neuroinflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Bianca Guglietti
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Srisankavi Sivasankar
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Sanam Mustafa
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia
| | - Frances Corrigan
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia. .,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
8
|
Bordone MP, Damianich A, Bernardi MA, Eidelman T, Sanz-Blasco S, Gershanik OS, Avale ME, Ferrario JE. Fyn knockdown prevents levodopa-induced dyskinesia in a mouse model of Parkinson's disease. eNeuro 2021; 8:ENEURO.0559-20.2021. [PMID: 34099487 PMCID: PMC8281260 DOI: 10.1523/eneuro.0559-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
Dopamine replacement by levodopa is the most widely used therapy for Parkinson's disease (PD), however patients often develop side effects, known as levodopa-induced dyskinesia (LID), that usually need therapeutic intervention. There are no suitable therapeutic options for LID, except for the use of the NMDA receptor antagonist amantadine, which has limited efficacy. The NMDA receptor is indeed the most plausible target to manage LID in PD and recently the kinase Fyn- one of its key regulators- became a new putative molecular target involved in LID. The aim of this work was to reduce Fyn expression to alleviate LID in a mouse model of PD. We performed intra-striatal delivery of a designed micro-RNA against Fyn (miRNA-Fyn) in 6-OHDA-lesioned mice treated with levodopa. The miRNA-Fyn was delivered either before or after levodopa exposure to assess its ability to prevent or revert dyskinesia. Pre-administration of miRNA-Fyn reduced LID with a concomitant reduction of FosB-ΔFosB protein levels -a marker of LID- as well as decreased phosphorylation of the NR2B-NMDA subunit, which is a main target of Fyn. On the other hand, post L-DOPA delivery of miRNA-Fyn was less effective to revert already established dyskinesia, suggesting that early blocking of Fyn activity might be a more efficient therapeutic approach. Together, our results provide proof of concept about Fyn as a plausible therapeutic target to manage LID, and validate RNA silencing as a potential approach to locally reduce striatal Fyn, rising new perspectives for RNA therapy interventions in PD.Significance StatementLevodopa induced dyskinesia (LID) is an incapacitant side effect of treatment in Parkinson's disease (PD). LID is a therapeutic challenge, lacking an effective pharmacological treatment, except for the use of inhibitors of the NMDA receptor, which have limited efficacy and may trigger untoward side effects. The kinase Fyn is a key regulator of NMDA function and a potential therapeutic target to control LID. Here, we show that RNA interference therapy to reduce the amount of Fyn mRNA in the adult brain is effective to prevent LID in a mouse model of PD, setting the grounds for future biomedical interventions to manage LID in PD.
Collapse
Affiliation(s)
- Melina P Bordone
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA)
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Ana Damianich
- CONICET - Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), "Dr. Héctor N. Torres", Ciudad Autónoma de Buenos Aires, Argentina (C1428ADN)
| | - M Alejandra Bernardi
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Tomas Eidelman
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA)
| | - Sara Sanz-Blasco
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Oscar S Gershanik
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - M Elena Avale
- CONICET - Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), "Dr. Héctor N. Torres", Ciudad Autónoma de Buenos Aires, Argentina (C1428ADN)
| | - Juan E Ferrario
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA).
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| |
Collapse
|
9
|
Xu J, Xue Y, Zhou R, Shi PY, Li H, Zhou J. Drug repurposing approach to combating coronavirus: Potential drugs and drug targets. Med Res Rev 2021; 41:1375-1426. [PMID: 33277927 PMCID: PMC8044022 DOI: 10.1002/med.21763] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/03/2020] [Accepted: 11/20/2020] [Indexed: 01/18/2023]
Abstract
In the past two decades, three highly pathogenic human coronaviruses severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus, and, recently, SARS-CoV-2, have caused pandemics of severe acute respiratory diseases with alarming morbidity and mortality. Due to the lack of specific anti-CoV therapies, the ongoing pandemic of coronavirus disease 2019 (COVID-19) poses a great challenge to clinical management and highlights an urgent need for effective interventions. Drug repurposing is a rapid and feasible strategy to identify effective drugs for combating this deadly infection. In this review, we summarize the therapeutic CoV targets, focus on the existing small molecule drugs that have the potential to be repurposed for existing and emerging CoV infections of the future, and discuss the clinical progress of developing small molecule drugs for COVID-19.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yu Xue
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
10
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
11
|
Gage MC, Thippeswamy T. Inhibitors of Src Family Kinases, Inducible Nitric Oxide Synthase, and NADPH Oxidase as Potential CNS Drug Targets for Neurological Diseases. CNS Drugs 2021; 35:1-20. [PMID: 33515429 PMCID: PMC7893831 DOI: 10.1007/s40263-020-00787-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/21/2022]
Abstract
Neurological diseases share common neuroinflammatory and oxidative stress pathways. Both phenotypic and molecular changes in microglia, astrocytes, and neurons contribute to the progression of disease and present potential targets for disease modification. Src family kinases (SFKs) are present in both neurons and glial cells and are upregulated following neurological insults in both human and animal models. In neurons, SFKs interact with post-synaptic protein domains to mediate hyperexcitability and neurotoxicity. SFKs are upstream of signaling cascades that lead to the modulation of neurotransmitter receptors and the transcription of pro-inflammatory cytokines as well as producers of free radicals through the activation of glia. Inducible nitric oxide synthase (iNOS/NOS-II) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), the major mediators of reactive nitrogen/oxygen species (RNS/ROS) production in the brain, are also upregulated along with the pro-inflammatory cytokines following neurological insult and contribute to disease progression. Persistent neuronal hyperexcitability, RNS/ROS, and cytokines can exacerbate neurodegeneration, a common pathognomonic feature of the most prevalent neurological disorders such as Alzheimer's disease, Parkinson's disease, and epilepsy. Using a wide variety of preclinical disease models, inhibitors of the SFK-iNOS-NOX2 signaling axis have been tested to cure or modify disease progression. In this review, we discuss the SFK-iNOS-NOX2 signaling pathway and their inhibitors as potential CNS targets for major neurological diseases.
Collapse
|
12
|
Pivotal Role of Fyn Kinase in Parkinson's Disease and Levodopa-Induced Dyskinesia: a Novel Therapeutic Target? Mol Neurobiol 2020; 58:1372-1391. [PMID: 33175322 DOI: 10.1007/s12035-020-02201-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
The exact etiology of Parkinson's disease (PD) remains obscure, although many cellular mechanisms including α-synuclein aggregation, oxidative damage, excessive neuroinflammation, and dopaminergic neuronal apoptosis are implicated in its pathogenesis. There is still no disease-modifying treatment for PD and the gold standard therapy, chronic use of levodopa is usually accompanied by severe side effects, mainly levodopa-induced dyskinesia (LID). Hence, the elucidation of the precise underlying molecular mechanisms is of paramount importance. Fyn is a tyrosine phospho-transferase of the Src family nonreceptor kinases that is highly implicated in immune regulation, cell proliferation and normal brain development. Accumulating preclinical evidence highlights the emerging role of Fyn in key aspects of PD and LID pathogenesis: it may regulate α-synuclein phosphorylation, oxidative stress-induced dopaminergic neuronal death, enhanced neuroinflammation and glutamate excitotoxicity by mediating key signaling pathways, such as BDNF/TrkB, PKCδ, MAPK, AMPK, NF-κB, Nrf2, and NMDAR axes. These findings suggest that therapeutic targeting of Fyn or Fyn-related pathways may represent a novel approach in PD treatment. Saracatinib, a nonselective Fyn inhibitor, has already been tested in clinical trials for Alzheimer's disease, and novel selective Fyn inhibitors are under investigation. In this comprehensive review, we discuss recent evidence on the role of Fyn in the pathogenesis of PD and LID and provide insights on additional Fyn-related molecular mechanisms to be explored in PD and LID pathology that could aid in the development of future Fyn-targeted therapeutic approaches.
Collapse
|
13
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
14
|
Luo X, Li Z, Zhao J, Deng Y, Zhong Y, Zhang M. Fyn gene silencing reduces oligodendrocytes apoptosis through inhibiting ERK1/2 phosphorylation in epilepsy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:298-304. [PMID: 31852295 DOI: 10.1080/21691401.2019.1671428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study aimed to investigate the effect of Fyn gene silencing on the apoptosis of oligodendrocytes (OLs) in epileptic model in vitro and the involved mechanism. Primary oligodendrocyte pro-genitor cells (OPCs) were separated from rats and differentiated to OLs. Immunofluorescent labeling showed positive expression of A2B5 in OPCs and Olig2 in OLs, suggesting the successful separation of OPCs and OLs. Three Fyn siRNAs (si-Fyn) and Fyn siRNA negative control (NC) were transfected into OLs. Western blot showed that among three si-Fyn groups, si-Fyn3 caused the lowest Fyn expression, so si-Fyn3 was chosen for following experiment. Cells were divided into four groups: Control, Model, NC and si-Fyn. In the Model group, cells were cultured in Mg-free extracellular fluid for 3 h. The morphology of control cells was normal. However, the migration of neurons, the aggregation of cell bodies and the "grid-like" changes of neural networks were observed in the model cells. OLs apoptosis in various groups was assessed by flow cytometry. Expression of Fyn, ERK1/2 and phosphorylated ERK1/2 (p-ERK1/2) in OLs of various groups was evaluated by western blot. Compared with the Control group, the apoptotic rates, the Fyn expression and p-ERK1/2/ERK1/2 ratio in the Model and NC groups increased significantly (p < .05). However, the apoptotic rate, the Fyn expression and p-ERK1/2/ERK1/2 ratio in the si-Fyn group were remarkably smaller than those in the Model group (p < .05). In conclusion, Fyn gene silencing reduced the apoptosis of OLs through inhibiting the phosphorylation of ERK1/2 in epileptic model.
Collapse
Affiliation(s)
- Xinming Luo
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhengyu Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhao
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Deng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuqin Zhong
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ming Zhang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Modified Glutamatergic Postsynapse in Neurodegenerative Disorders. Neuroscience 2019; 454:116-139. [PMID: 31887357 DOI: 10.1016/j.neuroscience.2019.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/02/2019] [Accepted: 12/02/2019] [Indexed: 01/27/2023]
Abstract
The postsynaptic density (PSD) is a complex subcellular domain important for postsynaptic signaling, function, and plasticity. The PSD is present at excitatory synapses and specialized to allow for precise neuron-to-neuron transmission of information. The PSD is localized immediately underneath the postsynaptic membrane forming a major protein network that regulates postsynaptic signaling and synaptic plasticity. Glutamatergic synaptic dysfunction affecting PSD morphology and signaling events have been described in many neurodegenerative disorders, either sporadic or familial forms. Thus, in this review we describe the main protein players forming the PSD and their activity, as well as relevant modifications in key components of the postsynaptic architecture occurring in Huntington's, Parkinson's and Alzheimer's diseases.
Collapse
|
16
|
Ravanidis S, Bougea A, Papagiannakis N, Maniati M, Koros C, Simitsi AM, Bozi M, Pachi I, Stamelou M, Paraskevas GP, Kapaki E, Moraitou M, Michelakakis H, Stefanis L, Doxakis E. Circulating Brain-enriched MicroRNAs for detection and discrimination of idiopathic and genetic Parkinson's disease. Mov Disord 2019; 35:457-467. [PMID: 31799764 DOI: 10.1002/mds.27928] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/13/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A minimally invasive test for early detection and monitoring of Parkinson's disease (PD) is a highly unmet need for drug development and planning of patient care. Blood plasma represents an attractive source of biomarkers. MicroRNAs (miRNAs) are conserved noncoding RNA molecules that serve as posttranscriptional regulators of gene expression. As opposed to ubiquitously expressed miRNAs that control house-keeping processes, brain-enriched miRNAs regulate diverse aspects of neuron development and function. These include neuron-subtype specification, axonal growth, dendritic morphogenesis, and spine density. Backed by a large number of studies, we now know that the differential expression of neuron-enriched miRNAs leads to brain dysfunction. OBJECTIVES The aim was to identify subsets of brain-enriched miRNAs with diagnostic potential for familial and idiopathic PD as well as specify the molecular pathways deregulated in PD. METHODS Initially, brain-enriched miRNAs were selected based on literature review and validation studies in human tissues. Subsequently, real-time reverse transcription polymerase chain reaction was performed in the plasma of 100 healthy controls and 99 idiopathic and 53 genetic (26 alpha-synucleinA53T and 27 glucocerebrosidase) patients. Statistical and bioinformatics analyses were carried out to pinpoint the diagnostic biomarkers and deregulated pathways, respectively. RESULTS An explicit molecular fingerprint for each of the 3 PD cohorts was generated. Although the idiopathic PD fingerprint was different from that of genetic PD, the molecular pathways deregulated converged between all PD subtypes. CONCLUSIONS The study provides a group of brain-enriched miRNAs that may be used for the detection and differentiation of PD subtypes. It has also identified the molecular pathways deregulated in PD. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Stylianos Ravanidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Anastasia Bougea
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Nikolaos Papagiannakis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Matina Maniati
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Christos Koros
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece.,2nd Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Maria Bozi
- 2nd Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioanna Pachi
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Maria Stamelou
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece.,Parkinson's disease and Movement disorders department, HYGEIA Hospital, Athens, Greece
| | - George P Paraskevas
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Elisabeth Kapaki
- 1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Marina Moraitou
- Department of Enzymology and Cellular Function, Institute of Child Health, Athens, Greece
| | - Helen Michelakakis
- Department of Enzymology and Cellular Function, Institute of Child Health, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,1st Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Epaminondas Doxakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
17
|
Eckernäs D, Hieronymus F, Carlsson T, Bergquist F. Acoustic white noise ameliorates reduced regional brain expression of CaMKII and ΔFosB in the spontaneously hypertensive rat model of ADHD. IBRO Rep 2019; 6:31-39. [PMID: 30656238 PMCID: PMC6302041 DOI: 10.1016/j.ibror.2018.11.007] [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: 08/01/2018] [Accepted: 11/28/2018] [Indexed: 11/29/2022] Open
Abstract
ΔFosB was reduced in the DL-PFC, DLS and nAc in SH rats. Acoustic noise normalized ΔFosB expression in the DL-PFC and nAc of SH rats. CaMKII expression was reduced in the TMN in SH rats. Acoustic noise increased CaMKII expression in the TMN in both strains.
Loud (≥70dBA) acoustic white noise improves cognitive performance in children with ADHD as well as skilled reach and rotarod performance in the spontaneously hypertensive (SH) rat model of ADHD. To investigate how acoustic noise influences the brain activity in the SH rat model of ADHD, immunohistochemical staining of two neuronal activity and plasticity markers, Ca2+/Calmodulin dependent protein kinase II (CaMKII) and ΔFosB, was evaluated in Wistar (n = 24) and SH (n = 16) rats after repeated exposure to acoustic noise or ambient silence. Other SH rats (n = 6) were treated with repeated methylphenidate (MPH). Expression of CaMKII was reduced in the tuberomammillary nucleus (TMN) of the SH rat compared to Wistar but not in the nucleus accumbens (nAc) or the dorsolateral prefrontal cortex (DL-PFC). In the TMN, the expression of CaMKII was increased by noise in both strains. ΔFosB expression was reduced in nAc, DL-PFC and the dorsolateral striatum (DLS) of the SH rat compared to Wistar. Exposure to acoustic white noise significantly increased ΔFosB expression in the nAc and DL-PFC but not in the DLS of SH rats. The results indicate that acoustic noise shifts a reduced neuronal activity in the nAc, TMN and DL-PFC in SH rats toward the normal levels of activity in outbred rats. This may explain why noise has benefit selectively in ADHD.
Collapse
Affiliation(s)
- Daniel Eckernäs
- Corresponding author at: University of Gothenburg, Institute of Neuroscience and Physiology, Department of Pharmacology, Box 431, 405 30, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
18
|
Ba M, Yu G, Yang H, Wang Y, Yu L, Kong M. Tat-Src reduced NR2B tyrosine phosphorylation and its interaction with NR2B in levodopa-induced dyskinetic rats model. Behav Brain Res 2018; 356:41-45. [PMID: 30130562 DOI: 10.1016/j.bbr.2018.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 12/27/2022]
Abstract
Augmented function of N-methyl-d-aspartate receptor subunit 2B (NR2B) and Src protein tyrosine kinase have been demonstrated to get involved in the pathological mechanisms of dyskinesia. In view of functional interactions between NR2B and Src, we investigated the effects of uncoupling NR2B and Src interactions on dyskinesia by using the Src-derived interfering peptide (Tat-Src). In the present study, valid 6-hydroxydopamine-lesioned parkinsonian rats were treated with levodopa intraperitoneally for 22 days to induce dyskinetic rats model. On day 23, dyskinetic rats received either Tat-Src or Tat-sSrc or vehicle with each levodopa dose. The data showed that in dyskinetic rats model intraperitoneal microinjection of Tat-Src improved dyskinetic behaviors and decreased NR2B tyrosine phosphorylation and the interactions of Src with NR2B induced by chronic levodopa treatment. Besides, Tat-Src also attenuated S-nitrosylation (SNO-Src) and the autophosphorylation (p-Src) of Src, which catalyzed NR2B phosphorylation. These findings suggest that targeting NR2B/Src complexes can be one potential treatment for dyskinesia in Parkinson's disease.
Collapse
Affiliation(s)
- Maowen Ba
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong 264000, PR China
| | - Guoping Yu
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong 264000, PR China
| | - Hongqi Yang
- Department of Neurology, Henan Provincial People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Ying Wang
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| | - Ling Yu
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China
| | - Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong 264000, PR China.
| |
Collapse
|