1
|
Göksu AY. A review article on the development of dopaminergic neurons and establishment of dopaminergic neuron-based in vitro models by using immortal cell lines or stem cells to study and treat Parkinson's disease. Int J Dev Neurosci 2024. [PMID: 39379284 DOI: 10.1002/jdn.10383] [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: 11/13/2023] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
The primary pathological hallmark of Parkinson's disease (PD) is the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta, a critical midbrain region. In vitro models based on DA neurons provide a powerful platform for investigating the cellular and molecular mechanisms of PD and testing novel therapeutic strategies. A deep understanding of DA neuron development, including the signalling pathways and transcription factors involved, is essential for advancing PD research. This article first explores the differentiation and maturation processes of DA neurons in the midbrain, detailing the relevant signalling pathways. It then compares various in vitro models, including primary cells, immortalized cell lines, and stem cell-based models, focusing on the advantages and limitations of each. Special attention is given to the role of immortalized and stem cell models in PD research. This review aims to guide researchers in selecting the most appropriate model for their specific research goals. Ethical considerations and clinical implications of using stem cells in PD research are also discussed.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and Embryology, Department of Gene and Cell Therapy, Akdeniz University, School of Medicine, Antalya, Turkey
| |
Collapse
|
2
|
Li S, Liu Y, Lu S, Xu J, Liu X, Yang D, Yang Y, Hou L, Li N. A crazy trio in Parkinson's disease: metabolism alteration, α-synuclein aggregation, and oxidative stress. Mol Cell Biochem 2024:10.1007/s11010-024-04985-3. [PMID: 38625515 DOI: 10.1007/s11010-024-04985-3] [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: 01/13/2024] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder, characterized by the progressive loss of dopaminergic neurons in the pars compacta of the substantia nigra and the presence of Lewy bodies containing α-synuclein within these neurons. Oligomeric α-synuclein exerts neurotoxic effects through mitochondrial dysfunction, glial cell inflammatory response, lysosomal dysfunction and so on. α-synuclein aggregation, often accompanied by oxidative stress, is generally considered to be a key factor in PD pathology. At present, emerging evidences suggest that metabolism alteration is closely associated with α-synuclein aggregation and PD progression, and improvement of key molecules in metabolism might be potentially beneficial in PD treatment. In this review, we highlight the tripartite relationship among metabolic changes, α-synuclein aggregation, and oxidative stress in PD, and offer updated insights into the treatments of PD, aiming to deepen our understanding of PD pathogenesis and explore new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sheng Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yanbing Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sen Lu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiayi Xu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaokun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Di Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ning Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
3
|
Zhao Y, Liu X, Yang G. Adenosinergic Pathway in Parkinson's Disease: Recent Advances and Therapeutic Perspective. Mol Neurobiol 2023; 60:3054-3070. [PMID: 36786912 DOI: 10.1007/s12035-023-03257-3] [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: 06/04/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized pathologically by α-synuclein (α-syn) aggregation. In PD, the current mainstay of symptomatic treatment is levodopa (L-DOPA)-based dopamine (DA) replacement therapy. However, the development of dyskinesia and/or motor fluctuations which is relevant to levodopa is restricting its long-term utility. Given that the ability of which is to modulate the striato-thalamo-cortical loops and function to modulate basal ganglia output, the adenosinergic pathway (AP) is qualified as a potential promising non-DA target. As an indispensable component of energy production pathways, AP modulates cellular metabolism and gene regulation in both neurons and neuroglia cells through the recognition and degradation of extracellular adenosine. In addition, AP is geared to the initiation, evolution, and resolution of inflammation as well. Besides the above-mentioned crosstalk between the adenosine and dopamine signaling pathways, the functions of adenosine receptors (A1R, A2AR, A2BR, and A3R) and metabolism enzymes in modulating PD pathological process have been extensively investigated in recent decades. Here we reviewed the emerging findings focused on the function of adenosine receptors, adenosine formation, and metabolism in the brain and discussed its potential roles in PD pathological process. We also recapitulated clinical studies and the preclinical evidence for the medical strategies targeting the Ado signaling pathway to improve motor dysfunction and alleviate pathogenic process in PD. We hope that further clinical studies should consider this pathway in their monotherapy and combination therapy, which would open new vistas to more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Xin Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Guofeng Yang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, People's Republic of China. .,Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
4
|
Waggan I, Rissanen E, Tuisku J, Joutsa J, Helin S, Parkkola R, Rinne JO, Airas L. Adenosine A 2A receptor availability in patients with early- and moderate-stage Parkinson's disease. J Neurol 2023; 270:300-310. [PMID: 36053386 DOI: 10.1007/s00415-022-11342-1] [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: 02/18/2022] [Revised: 08/06/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Adenosine 2A (A2A) receptors co-localize with dopamine D2 receptors in striatopallidal medium spiny neurons of the indirect pathway. A2A receptor activation in the striatum or pallidum decreases D2 signaling. In contrast, A2A receptor antagonism may help potentiate it. Furthermore, previous PET studies have shown increased A2A receptor availability in striatum of late-stage PD patients with dyskinesia. However, human in vivo evidence for striatal A2A receptor availability in early-stage PD is limited. This study aimed to investigate possible differences in A2A receptor availability in the striatum and pallidum of early- and moderate-stage PD patients without dyskinesias. METHODS Brain MRI and PET with [11C]TMSX radioligand, targeting A2A receptors, was performed in 9 patients with early- and 9 with moderate-stage PD without dyskinesia and in 6 healthy controls. Distribution volume ratios (DVR) were calculated to assess specific [11C]TMSX binding in caudate, putamen and pallidum. RESULTS A2A receptor availability (DVR) was decreased in the bilateral caudate of early-stage PD patients when compared with healthy controls (P = 0.02). Conversely, DVR was increased bilaterally in the pallidum of moderate-stage PD patients compared to healthy controls (P = 0.03). Increased mean striatal DVR correlated with higher motor symptom severity ([Formula: see text] = 0.47, P = 0.02). CONCLUSION Our results imply regional and disease stage-dependent changes in A2A receptor signaling in PD pathophysiology and in response to dopaminergic medication.
Collapse
Affiliation(s)
- Imran Waggan
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland.
| | - Eero Rissanen
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
| | - Juho Joutsa
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital and University of Turku, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Laura Airas
- Turku PET Centre, University of Turku, Itäinen Pitkäkatu 4A, 6th floor, 6007, 20520, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
5
|
Kondeva-Burdina M, Mitkov J, Valkova I, Peikova L, Georgieva M, Zlatkov A. Quantitative Structure-Neurotoxicity Assessment and In Vitro Evaluation of Neuroprotective and MAO-B Inhibitory Activities of Series N'-substituted 3-(1,3,7-trimethyl-xanthin-8-ylthio)propanehydrazides. Molecules 2022; 27:molecules27165321. [PMID: 36014559 PMCID: PMC9414684 DOI: 10.3390/molecules27165321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The neurotoxic, neuroprotective and MAO-B inhibitory effects of series N'-substituted 3-(1,3,7-trimethyl-xanthin-8-ylthio)propanehydrazides are evaluated. The results indicate compounds N'-(2,3-dimethoxybenzylidene)-3-(1,3,7-trimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-ylthio)propanehydrazide (6k) and N'-(2-hydroxybenzylidene)-3-(1,3,7-trimethyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-ylthio)propanehydrazide (6l) as most perspective. The performed QSTR analysis identified that the decreased lipophilicity and smaller dipole moments of the molecules are the structural features ensuring lower neurotoxicity. The obtained results may be used as initial information in the further design of (xanthinyl-8-ylthio)propanhydrazides with potential hMAOB inhibitory effect and pronounced neuroprotection.
Collapse
Affiliation(s)
- Magdalena Kondeva-Burdina
- Laboratory of Drug Metabolism and Drug Toxicity, Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
- Correspondence:
| | - Javor Mitkov
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
| | - Iva Valkova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
| | - Lily Peikova
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
| | - Maya Georgieva
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
| | - Alexander Zlatkov
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria
| |
Collapse
|
6
|
Transcriptome Profiling Reveals Differential Expression of Circadian Behavior Genes in Peripheral Blood of Monozygotic Twins Discordant for Parkinson's Disease. Cells 2022; 11:cells11162599. [PMID: 36010675 PMCID: PMC9406852 DOI: 10.3390/cells11162599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases. Investigating individuals with the most identical genetic background is optimal for minimizing the genetic contribution to gene expression. These individuals include monozygotic twins discordant for PD. Monozygotic twins have the same genetic background, age, sex, and often similar environmental conditions. The aim of this study was to carry out a transcriptome analysis of the peripheral blood of three pairs of monozygotic twins discordant for PD. We identified the metabolic process “circadian behavior” as a priority process for further study. Different expression of genes included in the term “circadian behavior” confirms that this process is involved in PD pathogenesis. We found increased expression of three genes associated with circadian behavior, i.e., PTGDS, ADORA2A, and MTA1, in twins with PD. These genes can be considered as potential candidate genes for this disease.
Collapse
|
7
|
Jakova E, Moutaoufik MT, Lee JS, Babu M, Cayabyab FS. Adenosine A1 receptor ligands bind to α-synuclein: implications for α-synuclein misfolding and α-synucleinopathy in Parkinson's disease. Transl Neurodegener 2022; 11:9. [PMID: 35139916 PMCID: PMC8830172 DOI: 10.1186/s40035-022-00284-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/21/2022] [Indexed: 12/20/2022] Open
Abstract
Background Accumulating α-synuclein (α-syn) aggregates in neurons and glial cells are the staples of many synucleinopathy disorders, such as Parkinson’s disease (PD). Since brain adenosine becomes greatly elevated in ageing brains and chronic adenosine A1 receptor (A1R) stimulation leads to neurodegeneration, we determined whether adenosine or A1R receptor ligands mimic the action of known compounds that promote α-syn aggregation (e.g., the amphetamine analogue 2-aminoindan) or inhibit α-syn aggregation (e.g., Rasagiline metabolite 1-aminoindan). In the present study, we determined whether adenosine, A1R receptor agonist N6-Cyclopentyladenosine (CPA) and antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) could directly interact with α-syn to modulate α-syn aggregation and neurodegeneration of dopaminergic neurons in the substantia nigra (SN). Methods Nanopore analysis and molecular docking were used to test the binding properties of CPA and DPCPX with α-syn in vitro. Sprague–Dawley rats were administered with 7-day intraperitoneal injections of the A1R ligands and 1- and 2-aminoindan, and levels of α-syn aggregation and neurodegeneration were examined in the SN pars compacta and hippocampal regions using confocal imaging and Western blotting. Results Using nanopore analysis, we showed that the A1R agonists (CPA and adenosine) interacted with the N-terminus of α-syn, similar to 2-aminoindan, which is expected to promote a “knot” conformation and α-syn misfolding. In contrast, the A1R antagonist DPCPX interacted with the N- and C-termini of α-syn, similar to 1-aminoindan, which is expected to promote a “loop” conformation that prevents α-syn misfolding. Molecular docking studies revealed that adenosine, CPA and 2-aminoindan interacted with the hydrophobic core of α-syn N-terminus, whereas DPCPX and 1-aminoindan showed direct binding to the N- and C-terminal hydrophobic pockets. Confocal imaging and Western blot analyses revealed that chronic treatments with CPA alone or in combination with 2-aminoindan increased α-syn expression/aggregation and neurodegeneration in both SN pars compacta and hippocampus. In contrast, DPCPX and 1-aminoindan attenuated the CPA-induced α-syn expression/aggregation and neurodegeneration in SN and hippocampus. Conclusions The results indicate that A1R agonists and drugs promoting a “knot” conformation of α-syn can cause α-synucleinopathy and increase neuronal degeneration, whereas A1R antagonists and drugs promoting a “loop” conformation of α-syn can be harnessed for possible neuroprotective therapies to decrease α-synucleinopathy in PD. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00284-3.
Collapse
Affiliation(s)
- Elisabet Jakova
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mohamed Taha Moutaoufik
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK, Canada
| | - Jeremy S Lee
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mohan Babu
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK, Canada
| | - Francisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
8
|
Gouda NA, Elkamhawy A, Cho J. Emerging Therapeutic Strategies for Parkinson’s Disease and Future Prospects: A 2021 Update. Biomedicines 2022; 10:biomedicines10020371. [PMID: 35203580 PMCID: PMC8962417 DOI: 10.3390/biomedicines10020371] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder pathologically distinguished by degeneration of dopaminergic neurons in the substantia nigra pars compacta. Muscle rigidity, tremor, and bradykinesia are all clinical motor hallmarks of PD. Several pathways have been implicated in PD etiology, including mitochondrial dysfunction, impaired protein clearance, and neuroinflammation, but how these factors interact remains incompletely understood. Although many breakthroughs in PD therapy have been accomplished, there is currently no cure for PD, only trials to alleviate the related motor symptoms. To reduce or stop the clinical progression and mobility impairment, a disease-modifying approach that can directly target the etiology rather than offering symptomatic alleviation remains a major unmet clinical need in the management of PD. In this review, we briefly introduce current treatments and pathophysiology of PD. In addition, we address the novel innovative therapeutic targets for PD therapy, including α-synuclein, autophagy, neurodegeneration, neuroinflammation, and others. Several immunomodulatory approaches and stem cell research currently in clinical trials with PD patients are also discussed. Moreover, preclinical studies and clinical trials evaluating the efficacy of novel and repurposed therapeutic agents and their pragmatic applications with encouraging outcomes are summarized. Finally, molecular biomarkers under active investigation are presented as potentially valuable tools for early PD diagnosis.
Collapse
Affiliation(s)
- Noha A. Gouda
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
| | - Ahmed Elkamhawy
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Jungsook Cho
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang 10326, Korea; (N.A.G.); (A.E.)
- Correspondence:
| |
Collapse
|
9
|
Franco R, Lillo A, Rivas-Santisteban R, Reyes-Resina I, Navarro G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021; 10:1124. [PMID: 34066933 PMCID: PMC8148598 DOI: 10.3390/cells10051124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal survival depends on the glia, that is, on the astroglial and microglial support. Neurons die and microglia are activated not only in neurodegenerative diseases but also in physiological aging. Activated microglia, once considered harmful, express two main phenotypes: the pro-inflammatory or M1, and the neuroprotective or M2. When neuroinflammation, i.e., microglial activation occurs, it is important to achieve a good M1/M2 balance, i.e., at some point M1 microglia must be skewed into M2 cells to impede chronic inflammation and to afford neuronal survival. G protein-coupled receptors in general and adenosine receptors in particular are potential targets for increasing the number of M2 cells. This article describes the mechanisms underlying microglial activation and analyzes whether these cells exposed to a first damaging event may be ready to be preconditioned to better react to exposure to more damaging events. Adenosine receptors are relevant due to their participation in preconditioning. They can also be overexpressed in activated microglial cells. The potential of adenosine receptors and complexes formed by adenosine receptors and cannabinoids as therapeutic targets to provide microglia-mediated neuroprotection is here discussed.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| | - Rafael Rivas-Santisteban
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| |
Collapse
|
10
|
Do caffeine and more selective adenosine A 2A receptor antagonists protect against dopaminergic neurodegeneration in Parkinson's disease? Parkinsonism Relat Disord 2020; 80 Suppl 1:S45-S53. [PMID: 33349580 PMCID: PMC8102090 DOI: 10.1016/j.parkreldis.2020.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/26/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022]
Abstract
The adenosine A2A receptor is a major target of caffeine, the most widely used psychoactive substance worldwide. Large epidemiological studies have long shown caffeine consumption is a strong inverse predictor of Parkinson’s disease (PD). In this review, we first examine the epidemiology of caffeine use vis-à-vis PD and follow this by looking at the evidence for adenosine A2A receptor antagonists as potential neuroprotective agents. There is a wealth of accumulating biological, epidemiological and clinical evidence to support the further investigation of selective adenosine A2A antagonists, as well as caffeine, as promising candidate therapeutics to fill the unmet need for disease modification of PD.
Collapse
|
11
|
Ren X, Chen JF. Caffeine and Parkinson's Disease: Multiple Benefits and Emerging Mechanisms. Front Neurosci 2020; 14:602697. [PMID: 33390888 PMCID: PMC7773776 DOI: 10.3389/fnins.2020.602697] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by dopaminergic neurodegeneration, motor impairment and non-motor symptoms. Epidemiological and experimental investigations into potential risk factors have firmly established that dietary factor caffeine, the most-widely consumed psychoactive substance, may exerts not only neuroprotective but a motor and non-motor (cognitive) benefits in PD. These multi-benefits of caffeine in PD are supported by convergence of epidemiological and animal evidence. At least six large prospective epidemiological studies have firmly established a relationship between increased caffeine consumption and decreased risk of developing PD. In addition, animal studies have also demonstrated that caffeine confers neuroprotection against dopaminergic neurodegeneration using PD models of mitochondrial toxins (MPTP, 6-OHDA, and rotenone) and expression of α-synuclein (α-Syn). While caffeine has complex pharmacological profiles, studies with genetic knockout mice have clearly revealed that caffeine’s action is largely mediated by the brain adenosine A2A receptor (A2AR) and confer neuroprotection by modulating neuroinflammation and excitotoxicity and mitochondrial function. Interestingly, recent studies have highlighted emerging new mechanisms including caffeine modulation of α-Syn degradation with enhanced autophagy and caffeine modulation of gut microbiota and gut-brain axis in PD models. Importantly, since the first clinical trial in 2003, United States FDA has finally approved clinical use of the A2AR antagonist istradefylline for the treatment of PD with OFF-time in Sept. 2019. To realize therapeutic potential of caffeine in PD, genetic study of caffeine and risk genes in human population may identify useful pharmacogenetic markers for predicting individual responses to caffeine in PD clinical trials and thus offer a unique opportunity for “personalized medicine” in PD.
Collapse
Affiliation(s)
- Xiangpeng Ren
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China.,Department of Biochemistry, Medical College, Jiaxing University, Jiaxing, China
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| |
Collapse
|
12
|
Glaser T, Andrejew R, Oliveira-Giacomelli Á, Ribeiro DE, Bonfim Marques L, Ye Q, Ren WJ, Semyanov A, Illes P, Tang Y, Ulrich H. Purinergic Receptors in Basal Ganglia Diseases: Shared Molecular Mechanisms between Huntington's and Parkinson's Disease. Neurosci Bull 2020; 36:1299-1314. [PMID: 33026587 PMCID: PMC7674528 DOI: 10.1007/s12264-020-00582-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/30/2020] [Indexed: 12/22/2022] Open
Abstract
Huntington's (HD) and Parkinson's diseases (PD) are neurodegenerative disorders caused by the death of GABAergic and dopaminergic neurons in the basal ganglia leading to hyperkinetic and hypokinetic symptoms, respectively. We review here the participation of purinergic receptors through intracellular Ca2+ signaling in these neurodegenerative diseases. The adenosine A2A receptor stimulates striatopallidal GABAergic neurons, resulting in inhibitory actions on GABAergic neurons of the globus pallidus. A2A and dopamine D2 receptors form functional heteromeric complexes inducing allosteric inhibition, and A2A receptor activation results in motor inhibition. Furthermore, the A2A receptor physically and functionally interacts with glutamate receptors, mainly with the mGlu5 receptor subtype. This interaction facilitates glutamate release, resulting in NMDA glutamate receptor activation and an increase of Ca2+ influx. P2X7 receptor activation also promotes glutamate release and neuronal damage. Thus, modulation of purinergic receptor activity, such as A2A and P2X7 receptors, and subsequent aberrant Ca2+ signaling, might present interesting therapeutic potential for HD and PD.
Collapse
Affiliation(s)
- Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Roberta Andrejew
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Ágatha Oliveira-Giacomelli
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Lucas Bonfim Marques
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Qing Ye
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil
- Key Laboratory of Sichuan Province for Acupuncture and Chronobiology, Chengdu, 610075, China
| | - Wen-Jing Ren
- Key Laboratory of Sichuan Province for Acupuncture and Chronobiology, Chengdu, 610075, China
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, 04107, Germany
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Sechenov First Moscow State Medical University, Moscow, 119992, Russia
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, 04107, Germany
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yong Tang
- Key Laboratory of Sichuan Province for Acupuncture and Chronobiology, Chengdu, 610075, China
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
13
|
Martí Navia A, Dal Ben D, Lambertucci C, Spinaci A, Volpini R, Marques-Morgado I, Coelho JE, Lopes LV, Marucci G, Buccioni M. Adenosine Receptors as Neuroinflammation Modulators: Role of A 1 Agonists and A 2A Antagonists. Cells 2020; 9:cells9071739. [PMID: 32708189 PMCID: PMC7409197 DOI: 10.3390/cells9071739] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/17/2020] [Indexed: 01/07/2023] Open
Abstract
The pathological condition of neuroinflammation is caused by the activation of the neuroimmune cells astrocytes and microglia. The autacoid adenosine seems to be an important neuromodulator in this condition. Its main receptors involved in the neuroinflammation modulation are A1AR and A2AAR. Evidence suggests that A1AR activation produces a neuroprotective effect and A2AARs block prevents neuroinflammation. The aim of this work is to elucidate the effects of these receptors in neuroinflammation using the partial agonist 2'-dCCPA (2-chloro-N6-cyclopentyl-2'-deoxyadenosine) (C1 KiA1AR = 550 nM, KiA2AAR = 24,800 nM, and KiA3AR = 5560 nM, α = 0.70, EC50A1AR = 832 nM) and the newly synthesized in house compound 8-chloro-9-ethyl-2-phenethoxyadenine (C2 KiA2AAR = 0.75 nM; KiA1AR = 17 nM and KiA3AR = 227 nM, IC50A2AAR = 251 nM unpublished results). The experiments were performed in in vitro and in in vivo models of neuroinflammation. Results showed that C1 was able to prevent the inflammatory effect induced by cytokine cocktail (TNF-α, IL-1β, and IFN-γ) while C2 possess both anti-inflammatory and antioxidant properties, counteracting both neuroinflammation in mixed glial cells and in an animal model of neuroinflammation. In conclusion, C2 is a potential candidate for neuroinflammation therapy.
Collapse
Affiliation(s)
- Aleix Martí Navia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| | - Inês Marques-Morgado
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Joana E. Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (I.M.-M.); (J.E.C.); (L.V.L.)
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
- Correspondence: ; Tel.: +39-073-7402-223
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy; (A.M.N.); (D.D.B.); (C.L.); (A.S.); (R.V.); (M.B.)
| |
Collapse
|
14
|
Schepici G, Silvestro S, Bramanti P, Mazzon E. Caffeine: An Overview of Its Beneficial Effects in Experimental Models and Clinical Trials of Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21134766. [PMID: 32635541 PMCID: PMC7369844 DOI: 10.3390/ijms21134766] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s Disease (PD) is a neurological disease characterized by the progressive degeneration of the nigrostriatal dopaminergic pathway with consequent loss of neurons in the substantia nigra pars compacta and dopamine depletion. The cytoplasmic inclusions of α-synuclein (α-Syn), known as Lewy bodies, are the cytologic hallmark of PD. The presence of α-Syn aggregates causes mitochondrial degeneration, responsible for the increase in oxidative stress and consequent neurodegeneration. PD is a progressive disease that shows a complicated pathogenesis. The current therapies are used to alleviate the symptoms of the disease without changing its clinical course. Recently, phytocompounds with neuroprotective effects and antioxidant properties such as caffeine have aroused the interest of researchers. The purpose of this review is to summarize the preclinical studies present in the literature and clinical trials recorded in ClinicalTrial.gov, aimed at illustrating the effects of caffeine used as a nutraceutical compound combined with the current PD therapies. Therefore, the preventive effects of caffeine in the neurodegeneration of dopaminergic neurons encourage the use of this alkaloid as a supplement to reduce the progress of the PD.
Collapse
|
15
|
Madeo G, Terraneo A, Cardullo S, Gómez Pérez LJ, Cellini N, Sarlo M, Bonci A, Gallimberti L. Long-Term Outcome of Repetitive Transcranial Magnetic Stimulation in a Large Cohort of Patients With Cocaine-Use Disorder: An Observational Study. Front Psychiatry 2020; 11:158. [PMID: 32180745 PMCID: PMC7059304 DOI: 10.3389/fpsyt.2020.00158] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/19/2020] [Indexed: 01/11/2023] Open
Abstract
Background: Cocaine is a psychostimulant drug used as performance enhancer throughout history. The prolonged use of cocaine is associated with addiction and a broad range of cognitive deficits. Currently, there are no medications proven to be effective for cocaine-use disorder (CocUD). Previous preliminary clinical work suggests some benefit from repetitive transcranial magnetic stimulation (rTMS) stimulating the prefrontal cortex (PFC), involved in inhibitory cognitive control, decision-making and attention. All published studies to date have been limited by small sample sizes and short follow-up times. Methods: This is a retrospective observational study of 284 outpatients (of whom 268 were men) meeting DSM-5 criteria for CocUD. At treatment entry, most were using cocaine every day or several times per week. All patients underwent 3 months of rTMS and were followed for up to 2 years, 8 months. Self-report, reports by family or significant others and regular urine screens were used to assess drug use. Results: Median time to the first lapse (resumption of cocaine use) since the beginning of treatment was 91 days. For most patients, TMS was re-administered weekly, then monthly, throughout follow-up. The decrease in frequency of rTMS sessions was not accompanied by an increase in lapses to cocaine use. Mean frequency of cocaine use was <1·0 day/month (median 0), while serious rTMS-related adverse events were infrequent, consistent with published reports from smaller studies. Conclusions: This is the first follow-up study to show that rTMS treatment is accompanied by long-lasting reductions in cocaine use in a large cohort.
Collapse
Affiliation(s)
| | | | | | | | - Nicola Cellini
- Department of General Psychology, University of Padua, Padua, Italy.,Padova Neuroscience Center, University of Padua, Padua, Italy
| | - Michela Sarlo
- Department of General Psychology, University of Padua, Padua, Italy.,Padova Neuroscience Center, University of Padua, Padua, Italy
| | | | | |
Collapse
|
16
|
Tóth A, Antal Z, Bereczki D, Sperlágh B. Purinergic Signalling in Parkinson's Disease: A Multi-target System to Combat Neurodegeneration. Neurochem Res 2019; 44:2413-2422. [PMID: 31054067 PMCID: PMC6776560 DOI: 10.1007/s11064-019-02798-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by progressive loss of dopaminergic neurons that results in characteristic motor and non-motor symptoms. L-3,4 dihydroxyphenylalanine (L-DOPA) is the gold standard therapy for the treatment of PD. However, long-term use of L-DOPA leads to side effects such as dyskinesias and motor fluctuation. Since purines have neurotransmitter and co-transmitter properties, the function of the purinergic system has been thoroughly studied in the nervous system. Adenosine and adenosine 5'-triphosphate (ATP) are modulators of dopaminergic neurotransmission, neuroinflammatory processes, oxidative stress, excitotoxicity and cell death via purinergic receptor subtypes. Aberrant purinergic receptor signalling can be either the cause or the result of numerous pathological conditions, including neurodegenerative disorders. Many data confirm the involvement of purinergic signalling pathways in PD. Modulation of purinergic receptor subtypes, the activity of ectonucleotidases and ATP transporters could be beneficial in the treatment of PD. We give a brief summary of the background of purinergic signalling focusing on its roles in PD. Possible targets for pharmacological treatment are highlighted.
Collapse
Affiliation(s)
- Adrián Tóth
- Department of Neurology, Faculty of Medicine, Semmelweis University, Balassa u. 6., Budapest, 1083, Hungary
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Üllői út 26., Budapest, 1085, Hungary
| | - Zsófia Antal
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary
| | - Dániel Bereczki
- Department of Neurology, Faculty of Medicine, Semmelweis University, Balassa u. 6., Budapest, 1083, Hungary
| | - Beáta Sperlágh
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary.
| |
Collapse
|
17
|
Borroto-Escuela DO, Fuxe K. Oligomeric Receptor Complexes and Their Allosteric Receptor-Receptor Interactions in the Plasma Membrane Represent a New Biological Principle for Integration of Signals in the CNS. Front Mol Neurosci 2019; 12:230. [PMID: 31607863 PMCID: PMC6773811 DOI: 10.3389/fnmol.2019.00230] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) not only exist as monomers but also as homomers and heteromers in which allosteric receptor-receptor interactions take place, modulating the functions of the participating GPCR protomers. GPCRs can also form heteroreceptor complexes with ionotropic receptors and receptor tyrosine kinases modulating their function. Furthermore, adaptor proteins interact with receptor protomers and modulate their interactions. The state of the art is that the allosteric receptor-receptor interactions are reciprocal, highly dynamic and substantially alter the signaling, trafficking, recognition and pharmacology of the participating protomers. The pattern of changes appears to be unique for each heteromer and can favor antagonistic or facilitatory interactions or switch the G protein coupling from e.g., Gi/o to Gq or to beta-arrestin signaling. It lends a new dimension to molecular integration in the nervous system. Future direction should be aimed at determining the receptor interface involving building models of selected heterodimers. This will make design of interface-interfering peptides that specifically disrupt the heterodimer possible. This will help to determine the functional role of the allosteric receptor-receptor interactions as well as the integration of signals at the plasma membrane by the heteroreceptor complexes, vs. integration of the intracellular signaling pathways. Integration of signals also at the plasma membrane seems crucial in view of the hypothesis that learning and memory at a molecular level takes place by reorganization of homo and heteroreceptor complexes in the postsynaptic membrane. Homo and heteroreceptor complexes are in balance with each other, and their disbalance is linked to disease. Targeting heteroreceptor complexes represents a novel strategy for the treatment of brain disorders.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, Urbino, Italy
- Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Franco R, Reyes-Resina I, Aguinaga D, Lillo A, Jiménez J, Raïch I, Borroto-Escuela DO, Ferreiro-Vera C, Canela EI, Sánchez de Medina V, Del Ser-Badia A, Fuxe K, Saura CA, Navarro G. Potentiation of cannabinoid signaling in microglia by adenosine A 2A receptor antagonists. Glia 2019; 67:2410-2423. [PMID: 31429130 DOI: 10.1002/glia.23694] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
Neuroprotective M2-skewed microglia appear as promising to alter the course of neurodegenerative diseases and G protein-coupled receptors (GPCRs) are potential targets to achieve such microglial polarization. A common feature of adenosine A2A (A2A R) and cannabinoid CB2 (CB2 R) GPCRs in microglia is that their expression is upregulated in Alzheimer's disease (AD). On the one hand, CB2 R seems a target for neuroprotection, delaying neurodegenerative processes like those associated to AD or Parkinson's diseases. A2A R antagonists reduce amyloid burden and improve cognitive performance and memory in AD animal models. We here show a close interrelationship between these two receptors in microglia; they are able to physically interact and affect the signaling of each other, likely due to conformational changes within the A2A -CB2 receptor heteromer (A2A -CB2 Het). Particularly relevant is the upregulation of A2A -CB2 Het expression in samples from the APPSw ,Ind AD transgenic mice model. The most relevant finding, confirmed in both heterologous cells and in primary cultures of microglia, was that blockade of A2A receptors results in increased CB2 R-mediated signaling. This heteromer-specific feature suggests that A2A R antagonists would potentiate, via microglia, the neuroprotective action of endocannabinoids with implications for AD therapy.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Reyes-Resina
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - David Aguinaga
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Lillo
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | - Jasmina Jiménez
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Iu Raïch
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | | | | | - Enric I Canela
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Anna Del Ser-Badia
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Carlos A Saura
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Investigating targets for neuropharmacological intervention by molecular dynamics simulations. Biochem Soc Trans 2019; 47:909-918. [PMID: 31085614 DOI: 10.1042/bst20190048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023]
Abstract
Medical research has identified over 500 brain disorders. Among these, there are still only very few neuropathologies whose causes are fully understood and, consequently, very few drugs whose mechanism of action is known. No FDA drug has been identified for major neurodegenerative diseases, such as Alzheimer's and Parkinson's. We still lack effective treatments and strategies for modulating progression or even early neurodegenerative disease onset diagnostic tools. A great support toward the highly needed identification of neuroactive drugs comes from computer simulation methods and, in particular, from molecular dynamics (MD). This provides insight into structure-function relationship of a target and predicts structure, dynamics and energetics of ligand/target complexes under biologically relevant conditions like temperature and physiological saline concentration. Here, we present examples of the predictive power of MD for neuroactive ligands/target complexes. This brief survey from our own research shows the usefulness of partnerships between academia and industry, and from joint efforts between experimental and theoretical groups.
Collapse
|
20
|
Alonso-Andrés P, Martín M, Albasanz JL. Modulation of Adenosine Receptors and Antioxidative Effect of Beer Extracts in in Vitro Models. Nutrients 2019; 11:nu11061258. [PMID: 31163630 PMCID: PMC6628356 DOI: 10.3390/nu11061258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
The fight against neurodegenerative diseases is promoting the searching of nutrients, preferably of wide consumption, with proven effects on health. Beer is widely consumed and has potential benefits on health. In this work, three different extracts from dark beer (DB), non-alcoholic beer (NAB), and lager beer (LB) were assayed at 30 min and 24 h in rat C6 glioma and human SH-SY5Y neuroblastoma cells in order to study their possible protective effects. Cell viability and adenosine A1, A2A, A2B, and A3 receptor gene expression and protein levels were measured in control cells and in cells challenged with hydrogen peroxide as an oxidant stressor. Among the three extracts analyzed, DB showed a greater protective effect against H2O2-induced oxidative stress and cell death. Moreover, a higher A1 receptor level was also induced by this extract. Interestingly, A1 receptor level was also increased by NAB and LB extracts, but to a lower extent, and the protective effect of these extracts against H2O2 was lower. This possible correlation between protection and A1 receptor level was observed at 24 h in both C6 and SH-SY5Y cells. In summary, different beer extracts modulate, to a different degree, adenosine receptors expression and protect both glioma and neuroblastoma cells from oxidative stress.
Collapse
Affiliation(s)
- Patricia Alonso-Andrés
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 13071 Ciudad Real, Spain.
| | - Mairena Martín
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 13071 Ciudad Real, Spain.
| | - José Luis Albasanz
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 13071 Ciudad Real, Spain.
| |
Collapse
|
21
|
Borroto-Escuela DO, Fuxe K. Adenosine heteroreceptor complexes in the basal ganglia are implicated in Parkinson's disease and its treatment. J Neural Transm (Vienna) 2019; 126:455-471. [PMID: 30637481 PMCID: PMC6456481 DOI: 10.1007/s00702-019-01969-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/06/2019] [Indexed: 02/08/2023]
Abstract
The adenosine homo, iso and heteroreceptor complexes in the basal ganglia play a highly significant role in modulating the indirect and direct pathways and the striosomal projections to the nigro-striatal DA system. The major adenosine receptor complexes in the striato-pallidal GABA neurons can be the A2AR-D2R and A2AR-D2R-mGluR5 receptor complexes, in which A2AR protomers and mGluR5 protomers can allosterically interact to inhibit D2R protomer signaling. Through a reorganization of these heteroreceptor complexes upon chronic dopaminergic treatment a pathological and prolonged inhibition of D2R receptor protomer signaling can develop with motor inhibition and wearing off of the therapeutic effects of levodopa and dopamine receptor agonists. The direct pathway is enriched in D1R in and around glutamate synapses enhancing the ability of these GABA neurons to be activated and increase motor initiation. The brake on these GABA neurons is in this case exerted by A1R forming A1R-D1R heteroreceptor complexes in which they allosterically inhibit D1R signaling and thereby reduce motor initiation. Upon chronic levodopa treatment a reorganization of the D1R heteroreceptor complexes develops with the formation of putative A1R-D1R-D3 in addition to D1R-D3R complexes in which D3R enhances D1R protomer signaling and may make the A1R protomer brake less effective. Alpha-synuclein monomers-dimers are postulated to form complexes with A2AR homo and heteroprotomers in the plasma membrane enhancing alpha-synuclein aggregation and toxicity. The alpha-synuclein fibrils formed in the A2AR enriched dendritic spines of the striato-pallidal GABA neurons may reach the surrounding DA terminals via extracellular-vesicle-mediated volume transmission involving internalization of the vesicles and their cargo (alpha-synuclein fibrils) into the vulnerable DA terminals, enhancing their degeneration followed by retrograde flow of these fibrils in the DA axons to the vulnerable nigral DA nerve cells.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Biomedicum, B0851, Solnavägen 9, 17177 Stockholm, Sweden
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100 Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Biomedicum, B0851, Solnavägen 9, 17177 Stockholm, Sweden
| |
Collapse
|
22
|
van Diggelen F, Hrle D, Apetri M, Christiansen G, Rammes G, Tepper A, Otzen DE. Two conformationally distinct α-synuclein oligomers share common epitopes and the ability to impair long-term potentiation. PLoS One 2019; 14:e0213663. [PMID: 30901378 PMCID: PMC6430514 DOI: 10.1371/journal.pone.0213663] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/26/2019] [Indexed: 02/01/2023] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disease for which there currently is no cure. Aggregation of the pre-synaptic protein α-synuclein (aSN) into oligomers (αSOs) is believed to play a key role in PD pathology, but little is known about αSO formation in vivo and how they induce neurodegeneration. Both the naturally occurring polyunsaturated fatty acid docosahexaenoic acid (DHA) and the lipid peroxidation product 4-hydroxynonenal (HNE), strongly upregulated during ROS conditions, stimulate the formation of αSOs, highlighting a potential role in PD. Yet, insight into αSOs structure and biological effects is still limited as most oligomer preparations studied to date are heterogeneous in composition. Here we have aggregated aSN in the presence of HNE and DHA and purified the αSOs using size exclusion chromatography. Both compounds stimulate formation of spherical αSOs containing anti-parallel β-sheet structure which have the same shape as unmodified αSOs though ca. 2-fold larger. Furthermore, the yield and stabilities of these oligomers are significantly higher than for unmodified aSN. Both modified and unmodified αSOs permeabilize synthetic vesicles, show high co-localisation with glutamatergic synapses and decrease Long Term Potentiation (LTP), in line with the reported synaptotoxic effects of αSOs. We conclude that DHA- and HNE-αSOs are convenient models for pathogenic disease-associated αSOs in PD.
Collapse
Affiliation(s)
- Femke van Diggelen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Crossbeta Biosciences BV, Utrecht, The Netherlands
| | - Dean Hrle
- Klinik für Anaesthesiologie der Technischen Universität München, Klinikum Rechts der Isar, Munich, Germany
| | | | | | - Gerhard Rammes
- Klinik für Anaesthesiologie der Technischen Universität München, Klinikum Rechts der Isar, Munich, Germany
| | | | - Daniel Erik Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- * E-mail:
| |
Collapse
|
23
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of Adenosine Receptors: The State of the Art. Physiol Rev 2018; 98:1591-1625. [PMID: 29848236 DOI: 10.1152/physrev.00049.2017] [Citation(s) in RCA: 475] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adenosine is a ubiquitous endogenous autacoid whose effects are triggered through the enrollment of four G protein-coupled receptors: A1, A2A, A2B, and A3. Due to the rapid generation of adenosine from cellular metabolism, and the widespread distribution of its receptor subtypes in almost all organs and tissues, this nucleoside induces a multitude of physiopathological effects, regulating central nervous, cardiovascular, peripheral, and immune systems. It is becoming clear that the expression patterns of adenosine receptors vary among cell types, lending weight to the idea that they may be both markers of pathologies and useful targets for novel drugs. This review offers an overview of current knowledge on adenosine receptors, including their characteristic structural features, molecular interactions and cellular functions, as well as their essential roles in pain, cancer, and neurodegenerative, inflammatory, and autoimmune diseases. Finally, we highlight the latest findings on molecules capable of targeting adenosine receptors and report which stage of drug development they have reached.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Gessi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Merighi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Fabrizio Vincenzi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Katia Varani
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| |
Collapse
|
24
|
Ghiglieri V, Calabrese V, Calabresi P. Alpha-Synuclein: From Early Synaptic Dysfunction to Neurodegeneration. Front Neurol 2018; 9:295. [PMID: 29780350 PMCID: PMC5945838 DOI: 10.3389/fneur.2018.00295] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/17/2018] [Indexed: 01/24/2023] Open
Abstract
Over the last two decades, many experimental and clinical studies have provided solid evidence that alpha-synuclein (α-syn), a small, natively unfolded protein, is closely related to Parkinson's disease (PD) pathology. To provide an overview on the different roles of this protein, here we propose a synopsis of seminal and recent studies that explored the many aspects of α-syn. Ranging from the physiological functions to its neurodegenerative potential, the relationship with the possible pathogenesis of PD will be discussed. Close attention will be paid on early cellular and molecular alterations associated with the presence of α-syn aggregates.
Collapse
Affiliation(s)
- Veronica Ghiglieri
- Dipartimento di Filosofia, Scienze Sociali, Umane e della Formazione, Università degli Studi di Perugia, Perugia, Italy.,Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Valeria Calabrese
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Paolo Calabresi
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, Rome, Italy.,Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia di Perugia, Perugia, Italy
| |
Collapse
|
25
|
Luan Y, Ren X, Zheng W, Zeng Z, Guo Y, Hou Z, Guo W, Chen X, Li F, Chen JF. Chronic Caffeine Treatment Protects Against α-Synucleinopathy by Reestablishing Autophagy Activity in the Mouse Striatum. Front Neurosci 2018; 12:301. [PMID: 29770111 PMCID: PMC5942142 DOI: 10.3389/fnins.2018.00301] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 04/18/2018] [Indexed: 12/31/2022] Open
Abstract
Despite converging epidemiological evidence for the inverse relationship of regular caffeine consumption and risk of developing Parkinson's disease (PD) with animal studies demonstrating protective effect of caffeine in various neurotoxin models of PD, whether caffeine can protect against mutant α-synuclein (α-Syn) A53T-induced neurotoxicity in intact animals has not been examined. Here, we determined the effect of chronic caffeine treatment using the α-Syn fibril model of PD by intra-striatal injection of preformed A53T α-Syn fibrils. We demonstrated that chronic caffeine treatment blunted a cascade of pathological events leading to α-synucleinopathy, including pSer129α-Syn-rich aggregates, apoptotic neuronal cell death, microglia, and astroglia reactivation. Importantly, chronic caffeine treatment did not affect autophagy processes in the normal striatum, but selectively reversed α-Syn-induced defects in macroautophagy (by enhancing microtubule-associated protein 1 light chain 3, and reducing the receptor protein sequestosome 1, SQSTM1/p62) and chaperone-mediated autophagy (CMA, by enhancing LAMP2A). These findings support that caffeine—a strongly protective environment factor as suggested by epidemiological evidence—may represent a novel pharmacological therapy for PD by targeting autophagy pathway.
Collapse
Affiliation(s)
- Yanan Luan
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiangpeng Ren
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Optometry and Vision Science, Wenzhou, China
| | - Wu Zheng
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Optometry and Vision Science, Wenzhou, China
| | - Zhenhai Zeng
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yingzi Guo
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhidong Hou
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei Guo
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Optometry and Vision Science, Wenzhou, China
| | - Xingjun Chen
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fei Li
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Optometry and Vision Science, Wenzhou, China.,Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
26
|
Sebastião AM, Rei N, Ribeiro JA. Amyotrophic Lateral Sclerosis (ALS) and Adenosine Receptors. Front Pharmacol 2018; 9:267. [PMID: 29713276 PMCID: PMC5911503 DOI: 10.3389/fphar.2018.00267] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
In the present review we discuss the potential involvement of adenosinergic signaling, in particular the role of adenosine receptors, in amyotrophic lateral sclerosis (ALS). Though the literature on this topic is not abundant, the information so far available on adenosine receptors in animal models of ALS highlights the interest to continue to explore the role of these receptors in this neurodegenerative disease. Indeed, all motor neurons affected in ALS are responsive to adenosine receptor ligands but interestingly, there are alterations in pre-symptomatic or early symptomatic stages that mirror those in advanced disease stages. Information starts to emerge pointing toward a beneficial role of A2A receptors (A2AR), most probably at early disease states, and a detrimental role of caffeine, in clear contrast with what occurs in other neurodegenerative diseases. However, some evidence also exists on a beneficial action of A2AR antagonists. It may happen that there are time windows where A2AR prove beneficial and others where their blockade is required. Furthermore, the same changes may not occur simultaneously at the different synapses. In line with this, it is not fully understood if ALS is a dying back disease or if it propagates in a centrifugal way. It thus seems crucial to understand how motor neuron dysfunction occurs, how adenosine receptors are involved in those dysfunctions and whether the early changes in purinergic signaling are compensatory or triggers for the disease. Getting this information is crucial before starting the design of purinergic based strategies to halt or delay disease progression.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
27
|
Oliveira-Giacomelli Á, Naaldijk Y, Sardá-Arroyo L, Gonçalves MCB, Corrêa-Velloso J, Pillat MM, de Souza HDN, Ulrich H. Purinergic Receptors in Neurological Diseases With Motor Symptoms: Targets for Therapy. Front Pharmacol 2018; 9:325. [PMID: 29692728 PMCID: PMC5902708 DOI: 10.3389/fphar.2018.00325] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 03/21/2018] [Indexed: 12/13/2022] Open
Abstract
Since proving adenosine triphosphate (ATP) functions as a neurotransmitter in neuron/glia interactions, the purinergic system has been more intensely studied within the scope of the central nervous system. In neurological disorders with associated motor symptoms, including Parkinson's disease (PD), motor neuron diseases (MND), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's Disease (HD), restless leg syndrome (RLS), and ataxias, alterations in purinergic receptor expression and activity have been noted, indicating a potential role for this system in disease etiology and progression. In neurodegenerative conditions, neural cell death provokes extensive ATP release and alters calcium signaling through purinergic receptor modulation. Consequently, neuroinflammatory responses, excitotoxicity and apoptosis are directly or indirectly induced. This review analyzes currently available data, which suggests involvement of the purinergic system in neuro-associated motor dysfunctions and underlying mechanisms. Possible targets for pharmacological interventions are also discussed.
Collapse
Affiliation(s)
| | - Yahaira Naaldijk
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura Sardá-Arroyo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Maria C. B. Gonçalves
- Department of Neurology and Neuroscience, Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana Corrêa-Velloso
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Micheli M. Pillat
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Héllio D. N. de Souza
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
Rajasundaram S. Adenosine A2A Receptor Signaling in the Immunopathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2018; 9:402. [PMID: 29559972 PMCID: PMC5845642 DOI: 10.3389/fimmu.2018.00402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/13/2018] [Indexed: 12/19/2022] Open
Abstract
Our increasing appreciation of adenosine as an endogenous signaling molecule that terminates inflammation has generated excitement regarding the potential to target adenosine receptors (ARs) in the treatment of multiple sclerosis (MS), a disease of chronic neuroinflammation. Of the four G protein-coupled ARs, A2ARs are the principal mediator of adenosine’s anti-inflammatory effects and accordingly, there is a growing body of evidence surrounding the role of A2ARs in experimental autoimmune encephalomyelitis (EAE), the dominant animal model of MS. Such evidence points to a complex, often paradoxical role for A2ARs in the immunopathogenesis of EAE, where they have the ability to both exacerbate and alleviate disease severity. This review seeks to interpret these paradoxical findings and evaluate the therapeutic promise of A2ARs. In essence, the complexities of A2AR signaling arise from two properties. Firstly, A2AR signaling downregulates the inflammatory potential of TH lymphocytes whilst simultaneously facilitating the recruitment of these cells into the CNS. Secondly, A2AR expression by myeloid cells – infiltrating macrophages and CNS-resident microglia – has the capacity to promote both tissue injury and repair in chronic neuroinflammation. Consequently, the therapeutic potential of targeting A2ARs is greatly undermined by the risk of collateral tissue damage in the periphery and/or CNS.
Collapse
|
29
|
Chen SQ, Wang ZS, Ma YX, Zhang W, Lu JL, Liang YR, Zheng XQ. Neuroprotective Effects and Mechanisms of Tea Bioactive Components in Neurodegenerative Diseases. Molecules 2018; 23:E512. [PMID: 29495349 PMCID: PMC6017384 DOI: 10.3390/molecules23030512] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 12/19/2022] Open
Abstract
As the population ages, neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD) impose a heavy burden on society and families. The pathogeneses of PD and AD are complex. There are no radical cures for the diseases, and existing therapeutic agents for PD and AD have diverse side effects. Tea contains many bioactive components such as polyphenols, theanine, caffeine, and theaflavins. Some investigations of epidemiology have demonstrated that drinking tea can decrease the risk of PD and AD. Tea polyphenols can lower the morbidity of PD and AD by reducing oxidative stress and regulating signaling pathways and metal chelation. Theanine can inhibit the glutamate receptors and regulate the extracellular concentration of glutamine, presenting neuroprotective effects. Additionally, the neuroprotective mechanisms of caffeine and theaflavins may contribute to the ability to antagonize the adenosine receptor A2AR and the antioxidant properties, respectively. Thus, tea bioactive components might be useful for neuronal degeneration treatment in the future. In the present paper, the neuro protection and the mechanisms of tea and its bioactive components are reviewed. Moreover, the potential challenges and future work are also discussed.
Collapse
Affiliation(s)
- Shu-Qing Chen
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Ze-Shi Wang
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Yi-Xiao Ma
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Wei Zhang
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Jian-Liang Lu
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Yue-Rong Liang
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| | - Xin-Qiang Zheng
- Tea Research Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.
| |
Collapse
|
30
|
Borroto-Escuela DO, Hinz S, Navarro G, Franco R, Müller CE, Fuxe K. Understanding the Role of Adenosine A2AR Heteroreceptor Complexes in Neurodegeneration and Neuroinflammation. Front Neurosci 2018; 12:43. [PMID: 29467608 PMCID: PMC5808169 DOI: 10.3389/fnins.2018.00043] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/17/2018] [Indexed: 11/13/2022] Open
Abstract
Adenosine is a nucleoside mainly formed by degradation of ATP, located intracellularly or extracellularly, and acts as a neuromodulator. It operates as a volume transmission signal through diffusion and flow in the extracellular space to modulate the activity of both glial cells and neurons. The effects of adenosine are mediated via four adenosine receptor subtypes: A1R, A2AR, A2BR, A3R. The A2AR has a wide-spread distribution but it is especially enriched in the ventral and dorsal striatum where it is mainly located in the striato-pallidal GABA neurons at a synaptic and extrasynaptic location. A number of A2AR heteroreceptor complexes exist in the striatum. The existence of A2AR-D2R heteroreceptor complexes with antagonistic A2AR-D2R interactions in the striato-pallidal GABA neurons is well-known with A2AR activation inhibiting Gi/o mediated signaling of D2Rs. A2AR-mGluR5 heteroreceptor complexes were also found in with synergistic receptor-receptor interactions enhancing the inhibition of the D2R protomer signaling. They are located mainly in extrasynaptic regions of the striato-pallidal GABA neurons. Results recently demonstrated the existence of brain A2AR-A2BR heteroreceptor complexes, in which A2BR protomer constitutively inhibited the function of the A2AR protomer. These adenosine A2AR heteroreceptor complexes may modulate alpha-synuclein aggregation and toxicity through postulated bidirectional direct interactions leading to marked increases in A2AR signaling both in nerve cells and microglia. It is of high interest that formation of A2AR-A2ABR heteroreceptor complexes provides a brake on A2AR recognition and signaling opening up a novel strategy for treatment of A2AR mediated neurodegeneration.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Physiology, Department of Biomolecular Science, University of Urbino, Campus Scientifico Enrico Mattei, Urbino, Italy
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Yaguajay, Cuba
| | - Sonja Hinz
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Rafael Franco
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
α-synuclein interacts with PrP C to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci 2017; 20:1569-1579. [PMID: 28945221 DOI: 10.1038/nn.4648] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Abstract
Synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies, are neurodegenerative disorders that are characterized by the accumulation of α-synuclein (aSyn) in intracellular inclusions known as Lewy bodies. Prefibrillar soluble aSyn oligomers, rather than larger inclusions, are currently considered to be crucial species underlying synaptic dysfunction. We identified the cellular prion protein (PrPC) as a key mediator in aSyn-induced synaptic impairment. The aSyn-associated impairment of long-term potentiation was blocked in Prnp null mice and rescued following PrPC blockade. We found that extracellular aSyn oligomers formed a complex with PrPC that induced the phosphorylation of Fyn kinase via metabotropic glutamate receptors 5 (mGluR5). aSyn engagement of PrPC and Fyn activated NMDA receptor (NMDAR) and altered calcium homeostasis. Blockade of mGluR5-evoked phosphorylation of NMDAR in aSyn transgenic mice rescued synaptic and cognitive deficits, supporting the hypothesis that a receptor-mediated mechanism, independent of pore formation and membrane leakage, is sufficient to trigger early synaptic damage induced by extracellular aSyn.
Collapse
|
32
|
Nrf2 activation by tauroursodeoxycholic acid in experimental models of Parkinson's disease. Exp Neurol 2017; 295:77-87. [DOI: 10.1016/j.expneurol.2017.05.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/05/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
|
33
|
Fazeli W, Zappettini S, Marguet SL, Grendel J, Esclapez M, Bernard C, Isbrandt D. Early-life exposure to caffeine affects the construction and activity of cortical networks in mice. Exp Neurol 2017; 295:88-103. [DOI: 10.1016/j.expneurol.2017.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 05/29/2017] [Indexed: 10/19/2022]
|
34
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pathological overproduction: the bad side of adenosine. Br J Pharmacol 2017; 174:1945-1960. [PMID: 28252203 PMCID: PMC6398520 DOI: 10.1111/bph.13763] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an endogenous ubiquitous purine nucleoside, which is increased by hypoxia, ischaemia and tissue damage and mediates a number of physiopathological effects by interacting with four GPCRs, identified as A1 , A2A , A2B and A3 . Physiological and acutely increased adenosine is mostly associated with beneficial effects that include vasodilatation and a decrease in inflammation. In contrast, chronic overproduction of adenosine occurs in important pathological states, where long-lasting increases in the nucleoside levels are responsible for the bad side of adenosine associated with chronic inflammation, fibrosis and organ damage. In this review, we describe and critically discuss the pathological overproduction of adenosine and analyse when, where and how adenosine exerts its detrimental effects throughout the body.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Gessi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Merighi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Fabrizio Vincenzi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Katia Varani
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| |
Collapse
|
35
|
Hurtado F, Cardenas MAN, Cardenas F, León LA. La Enfermedad de Parkinson: Etiología, Tratamientos y Factores Preventivos. UNIVERSITAS PSYCHOLOGICA 2017. [DOI: 10.11144/javeriana.upsy15-5.epet] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
La enfermedad de Parkinson (EP) es la patología neurodegenerativa motora con mayor incidencia a nivel mundial. Esta afecta a aproximadamente 2-3% de la población mayor a 60 años de edad y sus causas aún no han sido bien determinadas. Actualmente no existe cura para esta patología; sin embargo, es posible contar con diferentes tratamientos que permiten aliviar algunos de sus síntomas y enlentecer su curso. Estos tratamientos tienen como premisa contrarrestar los efectos ocasionados por la pérdida de la función dopaminérgica de la sustancia nigra (SN) sobre estructuras como el núcleo subtálamico (NST) o globo pálido interno (GPi) ya sea por medio de tratamientos farmacológicos, estimulación cerebral profunda (ECP) o con el implante celular. Existen también investigaciones que están dirigiendo su interés al desarrollo de fármacos con potencial terapéutico, que presenten alta especificidad a receptores colinérgicos de nicotina (nAChRs) y antagonistas de receptores de adenosina, específicamente del subtipo A2A. Estos últimos, juegan un papel importante en el control de liberación dopaminérgica y en los procesos de neuroprotección. En esta revisión se pretende ofrecer una panorámica actual sobre algunos de los factores de riesgo asociados a EP, algunos de los tratamientos actuales más utilizados y acerca del rol de sustancias potencialmente útiles en la prevención de esta enfermedad.
Collapse
|
36
|
Wilkaniec A, Gąssowska M, Czapski GA, Cieślik M, Sulkowski G, Adamczyk A. P2X7 receptor-pannexin 1 interaction mediates extracellular alpha-synuclein-induced ATP release in neuroblastoma SH-SY5Y cells. Purinergic Signal 2017; 13:347-361. [PMID: 28516276 PMCID: PMC5563296 DOI: 10.1007/s11302-017-9567-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/30/2017] [Indexed: 12/14/2022] Open
Abstract
Abnormalities of alpha-synuclein (ASN), the main component of protein deposits (Lewy bodies), were observed in Parkinson’s disease (PD), dementia with Lewy bodies, Alzheimer’s disease, and other neurodegenerative disorders. These alterations include increase in the levels of soluble ASN oligomers in the extracellular space. Numerous works have identified several mechanisms of their toxicity, including stimulation of the microglial P2X7 receptor leading to oxidative stress. While the significant role of purinergic signaling—particularly, P2 family receptors—in neurodegenerative disorders is well known, the interaction of extracellular soluble ASN with neuronal purinergic receptors is yet to be studied. Therefore, in this study, we have investigated the effect of ASN on P2 purinergic receptors and ATP-dependent signaling. We used neuroblastoma SH-SY5Y cell line and rat synaptoneurosomes treated with exogenous soluble ASN. The experiments were performed using spectrofluorometric, radiochemical, and immunochemical methods. We found the following: (i) ASN-induced intracellular free calcium mobilization in neuronal cells and nerve endings depends on the activation of purinergic P2X7 receptors; (ii) activation of P2X7 receptors leads to pannexin 1 recruitment to form an active complex responsible for ATP release; and (iii) ASN greatly decreases the activity of extracellular ecto-ATPase responsible for ATP degradation. Thus, it is concluded that purinergic receptors might be putative pharmacological targets in the molecular mechanism of extracellular ASN toxicity. Interference with P2X7 signaling seems to be a promising strategy for the prevention or therapy of PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland.
| | - Magdalena Gąssowska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland
| | - Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland
| | - Grzegorz Sulkowski
- Department of Neurochemistry, Laboratory of Pathoneurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 St., 02-106, Warsaw, Poland
| |
Collapse
|
37
|
Prolactin-induced neuroprotection against glutamate excitotoxicity is mediated by the reduction of [Ca2+]i overload and NF-κB activation. PLoS One 2017; 12:e0176910. [PMID: 28475602 PMCID: PMC5419567 DOI: 10.1371/journal.pone.0176910] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 04/19/2017] [Indexed: 12/16/2022] Open
Abstract
Prolactin (PRL) is a peptidic hormone that displays pleiotropic functions in the organism including different actions in the brain. PRL exerts a neuroprotective effect against excitotoxicity produced by glutamate (Glu) or kainic acid in both in vitro and in vivo models. It is well known that Glu excitotoxicity causes cell death through apoptotic or necrotic pathways due to intracellular calcium ([Ca2+] i) overload. Therefore, the aim of the present study was to assess the molecular mechanisms by which PRL maintains cellular viability of primary cultures of rat hippocampal neurons exposed to Glu excitotoxicity. We determined cell viability by monitoring mitochondrial activity and using fluorescent markers for viable and dead cells. The intracellular calcium level was determined by a fluorometric assay and proteins involved in the apoptotic pathway were determined by immunoblot. Our results demonstrated that PRL afforded neuroprotection against Glu excitotoxicity, as evidenced by a decrease in propidium iodide staining and by the decrease of the LDH activity. In addition, the MTT assay shows that PRL maintains normal mitochondrial activity even in neurons exposed to Glu. Furthermore, the Glu-induced intracellular [Ca2+]i overload was attenuated by PRL. These data correlate with the reduction found in the level of active caspase-3 and the pro-apoptotic ratio (Bax/Bcl-2). Concomitantly, PRL elicited the nuclear translocation of the transcriptional factor NF-κB, which was detected by immunofluorescence and confocal microscopy. To our knowledge, this is the first report demonstrating that PRL prevents Glu excitotoxicity by a mechanism involving the restoration of the intracellular calcium homeostasis and mitochondrial activity, as well as an anti-apoptotic action possibly mediated by the activity of NF-κB. Overall, the current results suggest that PRL could be of potential therapeutic advantage in the treatment of neurodegenerative diseases.
Collapse
|
38
|
Epigenetics in Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:363-390. [DOI: 10.1007/978-3-319-53889-1_19] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
39
|
Cunha RA. How does adenosine control neuronal dysfunction and neurodegeneration? J Neurochem 2016; 139:1019-1055. [PMID: 27365148 DOI: 10.1111/jnc.13724] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022]
Abstract
The adenosine modulation system mostly operates through inhibitory A1 (A1 R) and facilitatory A2A receptors (A2A R) in the brain. The activity-dependent release of adenosine acts as a brake of excitatory transmission through A1 R, which are enriched in glutamatergic terminals. Adenosine sharpens salience of information encoding in neuronal circuits: high-frequency stimulation triggers ATP release in the 'activated' synapse, which is locally converted by ecto-nucleotidases into adenosine to selectively activate A2A R; A2A R switch off A1 R and CB1 receptors, bolster glutamate release and NMDA receptors to assist increasing synaptic plasticity in the 'activated' synapse; the parallel engagement of the astrocytic syncytium releases adenosine further inhibiting neighboring synapses, thus sharpening the encoded plastic change. Brain insults trigger a large outflow of adenosine and ATP, as a danger signal. A1 R are a hurdle for damage initiation, but they desensitize upon prolonged activation. However, if the insult is near-threshold and/or of short-duration, A1 R trigger preconditioning, which may limit the spread of damage. Brain insults also up-regulate A2A R, probably to bolster adaptive changes, but this heightens brain damage since A2A R blockade affords neuroprotection in models of epilepsy, depression, Alzheimer's, or Parkinson's disease. This initially involves a control of synaptotoxicity by neuronal A2A R, whereas astrocytic and microglia A2A R might control the spread of damage. The A2A R signaling mechanisms are largely unknown since A2A R are pleiotropic, coupling to different G proteins and non-canonical pathways to control the viability of glutamatergic synapses, neuroinflammation, mitochondria function, and cytoskeleton dynamics. Thus, simultaneously bolstering A1 R preconditioning and preventing excessive A2A R function might afford maximal neuroprotection. The main physiological role of the adenosine modulation system is to sharp the salience of information encoding through a combined action of adenosine A2A receptors (A2A R) in the synapse undergoing an alteration of synaptic efficiency with an increased inhibitory action of A1 R in all surrounding synapses. Brain insults trigger an up-regulation of A2A R in an attempt to bolster adaptive plasticity together with adenosine release and A1 R desensitization; this favors synaptotocity (increased A2A R) and decreases the hurdle to undergo degeneration (decreased A1 R). Maximal neuroprotection is expected to result from a combined A2A R blockade and increased A1 R activation. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
40
|
Hu Q, Ren X, Liu Y, Li Z, Zhang L, Chen X, He C, Chen JF. Aberrant adenosine A2A receptor signaling contributes to neurodegeneration and cognitive impairments in a mouse model of synucleinopathy. Exp Neurol 2016; 283:213-23. [PMID: 27342081 DOI: 10.1016/j.expneurol.2016.05.040] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 02/08/2023]
Abstract
Synucleinopathy is characterized by abnormal accumulation of misfolded α-synuclein (α-Syn)-positive cytoplasmic inclusions and by neurodegeneration and cognitive impairments, but the pathogenesis mechanism of synucleinopathy remains to be defined. Using a transmission model of synucleinopathy by intracerebral injection of preformed A53T α-Syn fibrils, we investigated whether aberrant adenosine A2A receptor (A2AR) signaling contributed to pathogenesis of synucleinopathy. We demonstrated that intra-hippocampal injection of preformed mutant α-Syn fibrils triggered a striking and selective induction of A2AR expression which was closely co-localized with pSer129 α-Syn-rich inclusions in neurons and glial cells of hippocampus. Importantly, by abolishing aberrant A2AR signaling triggered by mutant α-Syn, genetic deletion of A2ARs blunted a cascade of pathological events leading to synucleinopathy, including pSer129 α-Syn-rich and p62-positive aggregates, NF-κB activation and astrogliosis, apoptotic neuronal cell death and working memory deficits without affecting motor activity. These findings define α-Syn-triggered aberrant A2AR signaling as a critical pathogenesis mechanism of synucleinopathy with dual controls of cognition and neurodegeneration by modulating α-Syn aggregates. Thus, aberrant A2AR signaling represents a useful biomarker as well as a therapeutic target of synucleinopathy.
Collapse
Affiliation(s)
- Qidi Hu
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangpeng Ren
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health, China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China.
| | - Ya Liu
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhihui Li
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health, China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Liping Zhang
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health, China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Xingjun Chen
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chaoxiang He
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health, China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China; Department of Neurology, Boston University School of Medicine, Boston, MA, United States.
| |
Collapse
|
41
|
Forloni G, Artuso V, La Vitola P, Balducci C. Oligomeropathies and pathogenesis of Alzheimer and Parkinson's diseases. Mov Disord 2016; 31:771-81. [DOI: 10.1002/mds.26624] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/13/2022] Open
Affiliation(s)
- Gianluigi Forloni
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| | | | - Pietro La Vitola
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| | - Claudia Balducci
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| |
Collapse
|
42
|
Fathalla AM, Soliman AM, Ali MH, Moustafa AA. Adenosine A2A Receptor Blockade Prevents Rotenone-Induced Motor Impairment in a Rat Model of Parkinsonism. Front Behav Neurosci 2016; 10:35. [PMID: 26973484 PMCID: PMC4770055 DOI: 10.3389/fnbeh.2016.00035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/15/2016] [Indexed: 12/12/2022] Open
Abstract
Pharmacological studies implicate the blockade of adenosine receptorsas an effective strategy for reducing Parkinson’s disease (PD) symptoms. The objective of this study is to elucidate the possible protective effects of ZM241385 and 8-cyclopentyl-1, 3-dipropylxanthine, two selective A2A and A1 receptor antagonists, on a rotenone rat model of PD. Rats were split into four groups: vehicle control (1 ml/kg/48 h), rotenone (1.5 mg/kg/48 h, s.c.), ZM241385 (3.3 mg/kg/day, i.p) and 8-cyclopentyl-1, 3-dipropylxanthine (5 mg/kg/day, i.p). After that, animals were subjected to behavioral (stride length and grid walking) and biochemical (measuring concentration of dopamine levels using high performance liquid chromatography, HPLC). In the rotenone group, rats displayed a reduced motor activity and disturbed movement coordination in the behavioral tests and a decreased dopamine concentration as foundby HPLC. The effect of rotenone was partially prevented in the ZM241385 group, but not with 8-cyclopentyl-1,3-dipropylxanthine administration. The administration of ZM241385 improved motor function and movement coordination (partial increase of stride length and partial decrease in the number of foot slips) and an increase in dopamine concentration in the rotenone-injected rats. However, the 8-cyclopentyl-1,3-dipropylxanthine and rotenone groups were not significantly different. These results indicate that selective A2A receptor blockade by ZM241385, but not A1 receptor blockadeby 8-cyclopentyl-1,3-dipropylxanthine, may treat PD motor symptoms. This reinforces the potential use of A2A receptor antagonists as a treatment strategy for PD patients.
Collapse
Affiliation(s)
- Ahmed M Fathalla
- Faculty of Medicine, Department of Pharmacology, Suez Canal University Ismailia, Egypt
| | - Amira M Soliman
- Faculty of Medicine, Department of Pharmacology, Suez Canal University Ismailia, Egypt
| | - Mohamed H Ali
- Center for Aging and Associated Diseases, Zewail City of Science and Technology Giza, Egypt
| | - Ahmed A Moustafa
- Department of Veterans Affairs, New Jersey Health Care SystemEast Orange, NJ, USA; School of Social Sciences and Psychology and Marcs Institute for Brain and Behaviour, Western Sydney UniversitySydney, NSW, Australia
| |
Collapse
|