1
|
Shields BC, Yan H, Lim SSX, Burwell SCV, Cammarata CM, Fleming EA, Yousefzadeh SA, Goldenshtein VZ, Kahuno EW, Vagadia PP, Loughran MH, Zhiquan L, McDonnell ME, Scalabrino ML, Thapa M, Hawley TM, Field GD, Hull C, Schiltz GE, Glickfeld LL, Reitz AB, Tadross MR. DART.2: bidirectional synaptic pharmacology with thousandfold cellular specificity. Nat Methods 2024; 21:1288-1297. [PMID: 38877316 DOI: 10.1038/s41592-024-02292-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/25/2024] [Indexed: 06/16/2024]
Abstract
Precision pharmacology aims to manipulate specific cellular interactions within complex tissues. In this pursuit, we introduce DART.2 (drug acutely restricted by tethering), a second-generation cell-specific pharmacology technology. The core advance is optimized cellular specificity-up to 3,000-fold in 15 min-enabling the targeted delivery of even epileptogenic drugs without off-target effects. Additionally, we introduce brain-wide dosing methods as an alternative to local cannulation and tracer reagents for brain-wide dose quantification. We describe four pharmaceuticals-two that antagonize excitatory and inhibitory postsynaptic receptors, and two that allosterically potentiate these receptors. Their versatility is showcased across multiple mouse-brain regions, including cerebellum, striatum, visual cortex and retina. Finally, in the ventral tegmental area, we find that blocking inhibitory inputs to dopamine neurons accelerates locomotion, contrasting with previous optogenetic and pharmacological findings. Beyond enabling the bidirectional perturbation of chemical synapses, these reagents offer intersectional precision-between genetically defined postsynaptic cells and neurotransmitter-defined presynaptic partners.
Collapse
Affiliation(s)
- Brenda C Shields
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Haidun Yan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Shaun S X Lim
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | | | | | | | | | - Purav P Vagadia
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | - Lei Zhiquan
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | | | - Mishek Thapa
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Tammy M Hawley
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Greg D Field
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Court Hull
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | - Allen B Reitz
- Fox Chase Therapeutics Discovery, Inc., Doylestown, PA, USA
| | - Michael R Tadross
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Xu YP, Zhang J, Mei X, Wu Y, Jiao W, Wang YH, Zhang AQ. Ablation of Shank1 Protects against 6-OHDA-induced Cytotoxicity via PRDX3-mediated Inhibition of ER Stress in SN4741 Cells. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:402-410. [PMID: 36797610 DOI: 10.2174/1871527322666230216124156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 02/18/2023]
Abstract
BACKGROUND Postsynaptic density (PSD) is an electron-dense structure that contains various scaffolding and signaling proteins. Shank1 is a master regulator of the synaptic scaffold located at glutamatergic synapses, and has been proposed to be involved in multiple neurological disorders. METHODS In this study, we investigated the role of shank1 in an in vitro Parkinson's disease (PD) model mimicked by 6-OHDA treatment in neuronal SN4741 cells. The expression of related molecules was detected by western blot and immunostaining. RESULTS We found that 6-OHDA significantly increased the mRNA and protein levels of shank1 in SN4741 cells, but the subcellular distribution was not altered. Knockdown of shank1 via small interfering RNA (siRNA) protected against 6-OHDA treatment, as evidenced by reduced lactate dehydrogenase (LDH) release and decreased apoptosis. The results of RT-PCR and western blot showed that knockdown of shank1 markedly inhibited the activation of endoplasmic reticulum (ER) stress associated factors after 6-OHDA exposure. In addition, the downregulation of shank1 obviously increased the expression of PRDX3, which was accompanied by the preservation of mitochondrial function. Mechanically, downregulation of PRDX3 via siRNA partially prevented the shank1 knockdowninduced protection against 6-OHDA in SN4741 cells. CONCLUSION In summary, the present study has provided the first evidence that the knockdown of shank1 protects against 6-OHDA-induced ER stress and mitochondrial dysfunction through activating the PRDX3 pathway.
Collapse
Affiliation(s)
- Ye-Ping Xu
- Department of Nursing, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210000, China
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
- Department of Nursing, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Jing Zhang
- Department of Nursing, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210000, China
- Department of Nursing, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Xue Mei
- Department of Nursing, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Yan Wu
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
- Department of Nursing, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Wei Jiao
- Department of Nursing, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Yu-Hai Wang
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Ai-Qin Zhang
- Department of Nursing, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
3
|
Serra M, Di Maio A, Bassareo V, Nuzzo T, Errico F, Servillo F, Capasso M, Parekh P, Li Q, Thiolat ML, Bezard E, Calabresi P, Sulzer D, Carta M, Morelli M, Usiello A. Perturbation of serine enantiomers homeostasis in the striatum of MPTP-lesioned monkeys and mice reflects the extent of dopaminergic midbrain degeneration. Neurobiol Dis 2023; 184:106226. [PMID: 37451474 DOI: 10.1016/j.nbd.2023.106226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Loss of dopaminergic midbrain neurons perturbs l-serine and d-serine homeostasis in the post-mortem caudate putamen (CPu) of Parkinson's disease (PD) patients. However, it is unclear whether the severity of dopaminergic nigrostriatal degeneration plays a role in deregulating serine enantiomers' metabolism. Here, through high-performance liquid chromatography (HPLC), we measured the levels of these amino acids in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys and MPTP-plus-probenecid (MPTPp)-treated mice to determine whether and how dopaminergic midbrain degeneration affects the levels of serine enantiomers in various basal ganglia subregions. In addition, in the same brain regions, we measured the levels of key neuroactive amino acids modulating glutamatergic neurotransmission, including l-glutamate, glycine, l-aspartate, d-aspartate, and their precursors l-glutamine, l-asparagine. In monkeys, MPTP treatment produced severe denervation of nigrostriatal dopaminergic fibers (⁓75%) and increased the levels of serine enantiomers in the rostral putamen (rPut), but not in the subthalamic nucleus, and the lateral and medial portion of the globus pallidus. Moreover, this neurotoxin significantly reduced the protein expression of the astrocytic serine transporter ASCT1 and the glycolytic enzyme GAPDH in the rPut of monkeys. Conversely, concentrations of d-serine and l-serine, as well as ASCT1 and GAPDH expression were unaffected in the striatum of MPTPp-treated mice, which showed only mild dopaminergic degeneration (⁓30%). These findings unveil a link between the severity of dopaminergic nigrostriatal degeneration and striatal serine enantiomers concentration, ASCT1 and GAPDH expression. We hypothesize that the up-regulation of d-serine and l-serine levels occurs as a secondary response within a homeostatic loop to support the metabolic and neurotransmission demands imposed by the degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Anna Di Maio
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Tommaso Nuzzo
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesco Errico
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy
| | - Federica Servillo
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy
| | - Mario Capasso
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini, 5, Napoli 80131, Italy
| | - Pathik Parekh
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Qin Li
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Marie-Laure Thiolat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Erwan Bezard
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China; Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Paolo Calabresi
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy; Neurologia, Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Alessandro Usiello
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
4
|
Di Maio A, Nuzzo T, Gilio L, Serra M, Buttari F, Errico F, De Rosa A, Bassi MS, Morelli M, Sasabe J, Sulzer D, Carta M, Centonze D, Usiello A. Homeostasis of serine enantiomers is disrupted in the post-mortem caudate putamen and cerebrospinal fluid of living Parkinson's disease patients. Neurobiol Dis 2023:106203. [PMID: 37336364 DOI: 10.1016/j.nbd.2023.106203] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
L-serine generated in astrocytes plays a pivotal role in modulating essential neurometabolic processes, while its enantiomer, D-serine, specifically regulates NMDA receptor (NMDAR) signalling. Despite their physiological relevance in modulating cerebral activity, serine enantiomers metabolism in Parkinson's disease (PD) remains elusive. Using High-Performance Liquid Chromatography (HPLC), we measured D- and L-serine levels along with other amino acids known to modulate NMDAR function, such as L-glutamate, L-aspartate, D-aspartate, and glycine, in the post-mortem caudate putamen (CPu) and superior frontal gyrus (SFG) of PD patients. Moreover, we examined these amino acids in the cerebrospinal fluid (CSF) of de novo living PD, Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) patients versus subjects with other neurological disorders (OND), used as control. We found higher D-serine and L-serine levels in the CPu of PD patients but not in the SFG, a cerebral region that, in contrast to the CPu, is not innervated by nigral dopaminergic terminals. We also highlighted a significant elevation of both serine enantiomers in the CSF samples from PD but not in those of AD and ALS patients, compared with control subjects. By contrast, none or only minor changes were found in the amount of other neuroactive amino acids mentioned above. Our findings identify D-serine and L-serine level upregulation as a biochemical signature associated with nigrostriatal dopaminergic degeneration in PD.
Collapse
Affiliation(s)
- Anna Di Maio
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Tommaso Nuzzo
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy; Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Francesco Errico
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy
| | - Arianna De Rosa
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | | | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Jumpei Sasabe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Alessandro Usiello
- Laboratory of Translational Neuroscience, Ceinge Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
5
|
Fan Y, Huang H, Shao J, Huang W. MicroRNA-mediated regulation of reactive astrocytes in central nervous system diseases. Front Mol Neurosci 2023; 15:1061343. [PMID: 36710937 PMCID: PMC9877358 DOI: 10.3389/fnmol.2022.1061343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Astrocytes (AST) are abundant glial cells in the human brain, accounting for approximately 20-50% percent of mammalian central nervous system (CNS) cells. They display essential functions necessary to sustain the physiological processes of the CNS, including maintaining neuronal structure, forming the blood-brain barrier, coordinating neuronal metabolism, maintaining the extracellular environment, regulating cerebral blood flow, stabilizing intercellular communication, participating in neurotransmitter synthesis, and defending against oxidative stress et al. During the pathological development of brain tumors, stroke, spinal cord injury (SCI), neurodegenerative diseases, and other neurological disorders, astrocytes undergo a series of highly heterogeneous changes, which are called reactive astrocytes, and mediate the corresponding pathophysiological process. However, the pathophysiological mechanisms of reactive astrocytes and their therapeutic relevance remain unclear. The microRNAs (miRNAs) are essential for cell differentiation, proliferation, and survival, which play a crucial role in the pathophysiological development of CNS diseases. In this review, we summarize the regulatory mechanism of miRNAs on reactive astrocytes in CNS diseases, which might provide a theoretical basis for the diagnosis and treatment of CNS diseases.
Collapse
|
6
|
Alijanpour S, Miryounesi M, Ghafouri-Fard S. The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders. Metab Brain Dis 2023; 38:1-16. [PMID: 36173507 DOI: 10.1007/s11011-022-01091-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/15/2022] [Indexed: 02/03/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). Excitatory amino acid transporters (EAATs) have important roles in the uptake of glutamate and termination of glutamatergic transmission. Up to now, five EAAT isoforms (EAAT1-5) have been identified in mammals. The main focus of this review is EAAT2. This protein has an important role in the pathoetiology of epilepsy. De novo dominant mutations, as well as inherited recessive mutation in this gene, have been associated with epilepsy. Moreover, dysregulation of this protein is implicated in a range of neurological diseases, namely amyotrophic lateral sclerosis, alzheimer's disease, parkinson's disease, schizophrenia, epilepsy, and autism. In this review, we summarize the role of EAAT2 in epilepsy and other neurological disorders, then provide an overview of the therapeutic modulation of this protein.
Collapse
Affiliation(s)
- Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Xie S, Yang J, Huang S, Fan Y, Xu T, He J, Guo J, Ji X, Wang Z, Li P, Chen J, Zhang Y. Disrupted myelination network in the cingulate cortex of Parkinson's disease. IET Syst Biol 2022; 16:98-119. [PMID: 35394697 PMCID: PMC9290774 DOI: 10.1049/syb2.12043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/31/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
The cingulate cortex is part of the conserved limbic system, which is considered as a hub of emotional and cognitive control. Accumulating evidence suggested that involvement of the cingulate cortex is significant for cognitive impairment of Parkinson's disease (PD). However, mechanistic studies of the cingulate cortex in PD pathogenesis are limited. Here, transcriptomic and regulatory network analyses were conducted for the cingulate cortex in PD. Enrichment and clustering analyses showed that genes involved in regulation of membrane potential and glutamate receptor signalling pathway were upregulated. Importantly, myelin genes and the oligodendrocyte development pathways were markedly downregulated, indicating disrupted myelination in PD cingulate cortex. Cell‐type‐specific signatures revealed that myelinating oligodendrocytes were the major cell type damaged in the PD cingulate cortex. Furthermore, downregulation of myelination pathways in the cingulate cortex were shared and validated in another independent RNAseq cohort of dementia with Lewy bodies (DLB). In combination with ATACseq data, gene regulatory networks (GRNs) were further constructed for 32 transcription factors (TFs) and 466 target genes among differentially expressed genes (DEGs) using a tree‐based machine learning algorithm. Several transcription factors, including Olig2, Sox8, Sox10, E2F1, and NKX6‐2, were highlighted as key nodes in a sub‐network, which control many overlapping downstream targets associated with myelin formation and gliogenesis. In addition, the authors have validated a subset of DEGs by qPCRs in two PD mouse models. Notably, seven of these genes,TOX3, NECAB2 NOS1, CAPN3, NR4A2, E2F1 and FOXP2, have been implicated previously in PD or neurodegeneration and are worthy of further studies as novel candidate genes. Together, our findings provide new insights into the role of remyelination as a promising new approach to treat PD after demyelination.
Collapse
Affiliation(s)
- Song Xie
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiajun Yang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shenghui Huang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuanlan Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tao Xu
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Jiangshuang He
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiahao Guo
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiang Ji
- Department of Mathematics, School of Science & Engineering, Tulane University, New Orleans, Louisiana, USA
| | - Zhibo Wang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Yi Zhang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China.,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
8
|
Ren C, He KJ, Hu H, Zhang JB, Dong LG, Li D, Chen J, Mao CJ, Wang F, Liu CF. Induction of Parkinsonian-Like Changes via Targeted Downregulation of Astrocytic Glutamate Transporter GLT-1 in the Striatum. JOURNAL OF PARKINSONS DISEASE 2021; 12:295-314. [PMID: 34719508 DOI: 10.3233/jpd-212640] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous investigations have suggested that decreased expression of glutamate transporter-1 (GLT-1) is involved in glutamate excitotoxicity and contribute to the development of Parkinson's disease (PD), GLT-1 is decreased in animal models of PD. GLT-1 is mainly expressed in astrocytes, and the striatum is a GLT-1-rich brain area. OBJECTIVE The aim was to explore the function and mechanism of astrocytic GLT-1 in PD-like changes. METHODS In the study, PD-like changes and their molecular mechanism in rodents were tested by a behavioral assessment, micro-positron emission tomography/computed tomography (PET/CT), western blotting, immunohistochemical and immunofluorescence staining, and high performance liquid chromatography pre-column derivatization with O-pthaldialdehida after downregulating astrocytic GLT-1 in vivo and in vitro. RESULTS In vivo, after 6 weeks of brain stereotactic injection of adeno-associated virus into the striatum, rats in the astrocytic GLT-1 knockdown group showed poorer motor performance, abnormal gait, and depression-like feature; but no olfactory disorders. The results of micro-PET/CT and western blotting indicated that the dopaminergic system was impaired in astrocytic GLT-1 knockdown rats. Similarly, tyrosine hydroxylase (TH) positive immune-staining in neurons of astrocytic GLT-1 knockdown rats showed deficit in cell count. In vitro, knockdown of astrocytic GLT-1 via RNA interference led to morphological injury of TH-positive neurons, which may be related to the abnormal calcium signal induced by glutamate accumulation after GLT-1 knockdown. Furthermore, the GLT-1 agonist ceftriaxone showed a protective effect on TH-positive neuron impairment. CONCLUSION The present findings may shed new light on the future prevention and treatment of PD based on blocking glutamate excitotoxicity.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kai-Jie He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hua Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dan Li
- Department of Neurology, Suqian First Hospital, Suqian, China
| | - Jing Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, Suqian First Hospital, Suqian, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China.,Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
9
|
The glutamatergic synapse: a complex machinery for information processing. Cogn Neurodyn 2021; 15:757-781. [PMID: 34603541 DOI: 10.1007/s11571-021-09679-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/04/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022] Open
Abstract
Being the most abundant synaptic type, the glutamatergic synapse is responsible for the larger part of the brain's information processing. Despite the conceptual simplicity of the basic mechanism of synaptic transmission, the glutamatergic synapse shows a large variation in the response to the presynaptic release of the neurotransmitter. This variability is observed not only among different synapses but also in the same single synapse. The synaptic response variability is due to several mechanisms of control of the information transferred among the neurons and suggests that the glutamatergic synapse is not a simple bridge for the transfer of information but plays an important role in its elaboration and management. The control of the synaptic information is operated at pre, post, and extrasynaptic sites in a sort of cooperation between the pre and postsynaptic neurons which also involves the activity of other neurons. The interaction between the different mechanisms of control is extremely complicated and its complete functionality is far from being fully understood. The present review, although not exhaustively, is intended to outline the most important of these mechanisms and their complexity, the understanding of which will be among the most intriguing challenges of future neuroscience.
Collapse
|
10
|
Meng X, Zhong J, Zeng C, Yung KKL, Zhang X, Wu X, Qu S. MiR-30a-5p Regulates GLT-1 Function via a PKCα-Mediated Ubiquitin Degradation Pathway in a Mouse Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:1578-1592. [PMID: 33882234 DOI: 10.1021/acschemneuro.1c00076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Glutamate excitotoxicity is caused by dysfunctional glutamate transporters and plays an important role in the pathogenesis of Parkinson's disease (PD); however, the mechanisms that underlie the regulation of glutamate transporters in PD are still not fully elucidated. MicroRNAs(miRNA), which are abundant in astrocytes and neurons, have been reported to play key roles in regulating the translation of glutamate-transporter mRNA. In this study, we hypothesized that the miR-30a-5p contributes to the pathogenesis of PD by regulating the ubiquitin-mediated degradation of glutamate transporter 1 (GLT-1). We demonstrated that short-hairpin RNA-mediated knockdown of miR-30a-5p ameliorated motor deficits and pathological changes like astrogliosis and reactive microgliosis in a mouse model of PD (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice). Western blotting and immunofluorescent labeling revealed that miR-30a-5p suppressed the expression and function of GLT-1 in MPTP-treated mice and specifically in astrocytes treated with 1-methyl-4-phenylpyridinium (MPP+) (cell model of PD). Both in vitro and in vivo, we found that miR-30a-5p knockdown promoted glutamate uptake and increased GLT-1 expression by hindering GLT-1 ubiquitination and subsequent degradation in a PKCα-dependent manner. Therefore, we conclude that miR-30a-5p represents a potential therapeutic target for the treatment of PD.
Collapse
Affiliation(s)
- Xingjun Meng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianping Zhong
- Department of Neurology, Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Chong Zeng
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaojuan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
11
|
Di Maio V, Santillo S, Ventriglia F. Synaptic dendritic activity modulates the single synaptic event. Cogn Neurodyn 2020; 15:279-297. [PMID: 33854645 DOI: 10.1007/s11571-020-09607-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 05/23/2020] [Accepted: 06/09/2020] [Indexed: 01/28/2023] Open
Abstract
Synaptic transmission is the key system for the information transfer and elaboration among neurons. Nevertheless, a synapse is not a standing alone structure but it is a part of a population of synapses inputting the information from several neurons on a specific area of the dendritic tree of a single neuron. This population consists of excitatory and inhibitory synapses the inputs of which drive the postsynaptic membrane potential in the depolarizing (excitatory synapses) or depolarizing (inhibitory synapses) direction modulating in such a way the postsynaptic membrane potential. The postsynaptic response of a single synapse depends on several biophysical factors the most important of which is the value of the membrane potential at which the response occurs. The concurrence in a specific time window of inputs by several synapses located in a specific area of the dendritic tree can, consequently, modulate the membrane potential such to severely influence the single postsynaptic response. The degree of modulation operated by the synaptic population depends on the number of synapses active, on the relative proportion between excitatory and inbibitory synapses belonging to the population and on their specific mean firing frequencies. In the present paper we show results obtained by the simulation of the activity of a single Glutamatergic excitatory synapse under the influence of two different populations composed of the same proportion of excitatory and inhibitory synapses but having two different sizes (total number of synapses). The most relevant conclusion of the present simulations is that the information transferred by the single synapse is not and independent simple transition between a pre- and a postsynaptic neuron but is the result of the cooperation of all the synapses which concurrently try to transfer the information to the postsynaptic neuron in a given time window. This cooperativeness is mainly operated by a simple mechanism of modulation of the postsynaptic membrane potential which influences the amplitude of the different components forming the postsynaptic excitatory response.
Collapse
Affiliation(s)
- Vito Di Maio
- Institute of Applied Science and Intelligent Systems (ISASI) of CNR, Pozzuoli, Italy
| | - Silvia Santillo
- Institute of Applied Science and Intelligent Systems (ISASI) of CNR, Pozzuoli, Italy
| | - Francesco Ventriglia
- Institute of Applied Science and Intelligent Systems (ISASI) of CNR, Pozzuoli, Italy
| |
Collapse
|
12
|
Synaptic GluN2A-Containing NMDA Receptors: From Physiology to Pathological Synaptic Plasticity. Int J Mol Sci 2020; 21:ijms21041538. [PMID: 32102377 PMCID: PMC7073220 DOI: 10.3390/ijms21041538] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-d-Aspartate Receptors (NMDARs) are ionotropic glutamate-gated receptors. NMDARs are tetramers composed by several homologous subunits of GluN1-, GluN2-, or GluN3-type, leading to the existence in the central nervous system of a high variety of receptor subtypes with different pharmacological and signaling properties. NMDAR subunit composition is strictly regulated during development and by activity-dependent synaptic plasticity. Given the differences between GluN2 regulatory subunits of NMDAR in several functions, here we will focus on the synaptic pool of NMDARs containing the GluN2A subunit, addressing its role in both physiology and pathological synaptic plasticity as well as the contribution in these events of different types of GluN2A-interacting proteins.
Collapse
|
13
|
The levels of the NMDA receptor co-agonist D-serine are reduced in the substantia nigra of MPTP-lesioned macaques and in the cerebrospinal fluid of Parkinson's disease patients. Sci Rep 2019; 9:8898. [PMID: 31222058 PMCID: PMC6586824 DOI: 10.1038/s41598-019-45419-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/03/2019] [Indexed: 01/31/2023] Open
Abstract
Dysfunction of NMDA receptor (NMDAR)-mediated transmission is supposed to contribute to the motor and non-motor symptoms of Parkinson’s Disease (PD), and to L-DOPA-induced dyskinesia. Besides the main agonist L-glutamate, two other amino acids in the atypical D-configuration, D-serine and D-aspartate, activate NMDARs. In the present work, we investigated the effect of dopamine depletion on D-amino acids metabolism in the brain of MPTP-lesioned Macaca mulatta, and in the serum and cerebrospinal fluid of PD patients. We found that MPTP treatment increases D-aspartate and D-serine in the monkey putamen while L-DOPA rescues both D-amino acids levels. Conversely, dopaminergic denervation is associated with selective D-serine reduction in the substantia nigra. Such decrease suggests that the beneficial effect of D-serine adjuvant therapy previously reported in PD patients may derive from the normalization of endogenous D-serine levels and consequent improvement of nigrostriatal hypoglutamatergic transmission at glycine binding site. We also found reduced D-serine concentration in the cerebrospinal fluid of L-DOPA-free PD patients. These results further confirm the existence of deep interaction between dopaminergic and glutamatergic neurotransmission in PD and disclose a possible direct influence of D-amino acids variations in the changes of NMDAR transmission occurring under dopamine denervation and L-DOPA therapy.
Collapse
|
14
|
Zhang J, Feng J, Ma D, Wang F, Wang Y, Li C, Wang X, Yin X, Zhang M, Dagda RK, Zhang Y. Neuroprotective Mitochondrial Remodeling by AKAP121/PKA Protects HT22 Cell from Glutamate-Induced Oxidative Stress. Mol Neurobiol 2019; 56:5586-5607. [PMID: 30652267 DOI: 10.1007/s12035-018-1464-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Protein kinase A (PKA) is a ser/thr kinase that is critical for maintaining essential neuronal functions including mitochondrial homeostasis, bioenergetics, neuronal development, and neurotransmission. The endogenous pool of PKA is targeted to the mitochondrion by forming a complex with the mitochondrial scaffold A-kinase anchoring protein 121 (AKAP121). Enhanced PKA signaling via AKAP121 leads to PKA-mediated phosphorylation of the fission modulator Drp1, leading to enhanced mitochondrial networks and thereby blocking apoptosis against different toxic insults. In this study, we show for the first time that AKAP121/PKA confers neuroprotection in an in vitro model of oxidative stress induced by exposure to excess glutamate. Unexpectedly, treating mouse hippocampal progenitor neuronal HT22 cells with an acute dose or chronic exposure of glutamate robustly elevates PKA signaling, a beneficial compensatory response that is phenocopied in HT22 cells conditioned to thrive in the presence of excess glutamate but not in parental HT22 cells. Secondly, redirecting the endogenous pool of PKA by transiently transfecting AKAP121 or transfecting a constitutively active mutant of PKA targeted to the mitochondrion (OMM-PKA) or of an isoform of AKAP121 that lacks the KH and Tudor domains (S-AKAP84) are sufficient to significantly block cell death induced by glutamate toxicity but not in an oxygen deprivation/reperfusion model. Conversely, transient transfection of HT22 neuronal cells with a PKA-binding-deficient mutant of AKAP121 is unable to protect against oxidative stress induced by glutamate toxicity suggesting that the catalytic activity of PKA is required for AKAP121's protective effects. Mechanistically, AKAP121 promotes neuroprotection by enhancing PKA-mediated phosphorylation of Drp1 to increase mitochondrial fusion, elevates ATP levels, and elicits an increase in the levels of antioxidants GSH and superoxide dismutase 2 leading to a reduction in the level of mitochondrial superoxide. Overall, our data supports AKAP121/PKA as a new molecular target that confers neuroprotection against glutamate toxicity by phosphorylating Drp1, to stabilize mitochondrial networks and mitochondrial function and to elicit antioxidant responses.
Collapse
Affiliation(s)
- Jingdian Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Feng Wang
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yumeng Wang
- Department of Physiology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Xinmin Street No. 126, Changchun, 130000, China
| | - Chunxiao Li
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Ruben K Dagda
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Mailstop 318, Howard Medical Sciences Building 148A (Office), Reno, NV, 89557,, USA
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China.
| |
Collapse
|
15
|
Mellone M, Zianni E, Stanic J, Campanelli F, Marino G, Ghiglieri V, Longhi A, Thiolat ML, Li Q, Calabresi P, Bezard E, Picconi B, Di Luca M, Gardoni F. NMDA receptor GluN2D subunit participates to levodopa-induced dyskinesia pathophysiology. Neurobiol Dis 2019; 121:338-349. [DOI: 10.1016/j.nbd.2018.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/11/2018] [Accepted: 09/23/2018] [Indexed: 12/17/2022] Open
|
16
|
Di Maio V, Santillo S, Ventriglia F. Multisynaptic cooperation shapes single glutamatergic synapse response. Brain Res 2018; 1697:93-104. [PMID: 29913131 DOI: 10.1016/j.brainres.2018.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 01/18/2023]
Abstract
The activity of thousands of excitatory synapse in the dendritic tree produces variations of membrane potential which, while can produce the spike generation at soma (hillock), can also influence the output of a single glutamatergic synapse. We used a model of synaptic diffusion and EPSP generation to simulate the effect of different number of active synapses on the output of a single one. Our results show that, also in subthreshold conditions, the excitatory dendritic activity can influence several parameters of the single synaptic output such as its amplitude, its time course, the NMDA-component activation and consequently phenomena like STP and LTP.
Collapse
Affiliation(s)
- Vito Di Maio
- Istituto di Scienze Applicate e Sistemi Intelligenti (ISASI) del CNR, Italy.
| | - Silvia Santillo
- Istituto di Scienze Applicate e Sistemi Intelligenti (ISASI) del CNR, Italy
| | | |
Collapse
|
17
|
Shields BC, Kahuno E, Kim C, Apostolides PF, Brown J, Lindo S, Mensh BD, Dudman JT, Lavis LD, Tadross MR. Deconstructing behavioral neuropharmacology with cellular specificity. Science 2017; 356:356/6333/eaaj2161. [PMID: 28385956 DOI: 10.1126/science.aaj2161] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/06/2017] [Indexed: 12/17/2022]
Abstract
Behavior has molecular, cellular, and circuit determinants. However, because many proteins are broadly expressed, their acute manipulation within defined cells has been difficult. Here, we combined the speed and molecular specificity of pharmacology with the cell type specificity of genetic tools. DART (drugs acutely restricted by tethering) is a technique that rapidly localizes drugs to the surface of defined cells, without prior modification of the native target. We first developed an AMPAR antagonist DART, with validation in cultured neuronal assays, in slices of mouse dorsal striatum, and in behaving mice. In parkinsonian animals, motor deficits were causally attributed to AMPARs in indirect spiny projection neurons (iSPNs) and to excess phasic firing of tonically active interneurons (TANs). Together, iSPNs and TANs (i.e., D2 cells) drove akinesia, whereas movement execution deficits reflected the ratio of AMPARs in D2 versus D1 cells. Finally, we designed a muscarinic antagonist DART in one iteration, demonstrating applicability of the method to diverse targets.
Collapse
Affiliation(s)
- Brenda C Shields
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Elizabeth Kahuno
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Charles Kim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Pierre F Apostolides
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Jennifer Brown
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Sarah Lindo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Brett D Mensh
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Joshua T Dudman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Michael R Tadross
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
18
|
Stanic J, Mellone M, Napolitano F, Racca C, Zianni E, Minocci D, Ghiglieri V, Thiolat ML, Li Q, Longhi A, De Rosa A, Picconi B, Bezard E, Calabresi P, Di Luca M, Usiello A, Gardoni F. Rabphilin 3A: A novel target for the treatment of levodopa-induced dyskinesias. Neurobiol Dis 2017; 108:54-64. [PMID: 28823933 DOI: 10.1016/j.nbd.2017.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/19/2017] [Accepted: 08/16/2017] [Indexed: 11/26/2022] Open
Abstract
N-methyl-d-aspartate receptor (NMDAR) subunit composition strictly commands receptor function and pharmacological responses. Changes in NMDAR subunit composition have been documented in brain disorders such as Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesias (LIDs), where an increase of NMDAR GluN2A/GluN2B subunit ratio at striatal synapses has been observed. A therapeutic approach aimed at rebalancing NMDAR synaptic composition represents a valuable strategy for PD and LIDs. To this, the comprehension of the molecular mechanisms regulating the synaptic localization of different NMDAR subtypes is required. We have recently demonstrated that Rabphilin 3A (Rph3A) is a new binding partner of NMDARs containing the GluN2A subunit and that it plays a crucial function in the synaptic stabilization of these receptors. Considering that protein-protein interactions govern the synaptic retention of NMDARs, the purpose of this work was to analyse the role of Rph3A and Rph3A/NMDAR complex in PD and LIDs, and to modulate Rph3A/GluN2A interaction to counteract the aberrant motor behaviour associated to chronic L-DOPA administration. Thus, an array of biochemical, immunohistochemical and pharmacological tools together with electron microscopy were applied in this study. Here we found that Rph3A is localized at the striatal postsynaptic density where it interacts with GluN2A. Notably, Rph3A expression at the synapse and its interaction with GluN2A-containing NMDARs were increased in parkinsonian rats displaying a dyskinetic profile. Acute treatment of dyskinetic animals with a cell-permeable peptide able to interfere with Rph3A/GluN2A binding significantly reduced their abnormal motor behaviour. Altogether, our findings indicate that Rph3A activity is linked to the aberrant synaptic localization of GluN2A-expressing NMDARs characterizing LIDs. Thus, we suggest that Rph3A/GluN2A complex could represent an innovative therapeutic target for those pathological conditions where NMDAR composition is significantly altered.
Collapse
Affiliation(s)
- Jennifer Stanic
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy
| | - Manuela Mellone
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania, Luigi Vanvitelli, Caserta, Italy
| | - Francesco Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Claudia Racca
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Elisa Zianni
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy
| | - Daiana Minocci
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy
| | - Veronica Ghiglieri
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, 00143 Roma, Italy; Department of Philosophy, Human, Social and Educational Sciences, University of Perugia, Perugia, Italy
| | - Marie-Laure Thiolat
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Qin Li
- Motac Neuroscience Ltd, Manchester, United Kingdom; Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Annalisa Longhi
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy
| | | | - Barbara Picconi
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, 00143 Roma, Italy
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, United Kingdom; Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Paolo Calabresi
- Laboratorio di Neurofisiologia, Fondazione Santa Lucia, IRCCS, 00143 Roma, Italy; Clinica Neurologica, Università degli studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia, Italy
| | - Monica Di Luca
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy
| | - Alessandro Usiello
- Ceinge Biotecnologie Avanzate, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania, Luigi Vanvitelli, Caserta, Italy
| | - Fabrizio Gardoni
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133, Milano, Italy.
| |
Collapse
|
19
|
Sassone J, Serratto G, Valtorta F, Silani V, Passafaro M, Ciammola A. The synaptic function of parkin. Brain 2017; 140:2265-2272. [PMID: 28335015 DOI: 10.1093/brain/awx006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Loss of function mutations in the gene PARK2, which encodes the protein parkin, cause autosomal recessive juvenile parkinsonism, a neurodegenerative disease characterized by degeneration of the dopaminergic neurons localized in the substantia nigra pars compacta. No therapy is effective in slowing disease progression mostly because the pathogenesis of the disease is yet to be understood. From accruing evidence suggesting that the protein parkin directly regulates synapses it can be hypothesized that PARK2 gene mutations lead to early synaptic damage that results in dopaminergic neuron loss over time. We review evidence that supports the role of parkin in modulating excitatory and dopaminergic synapse functions. We also discuss how these findings underpin the concept that autosomal recessive juvenile parkinsonism can be primarily a synaptopathy. Investigation into the molecular interactions between parkin and synaptic proteins may yield novel targets for pharmacologic interventions.
Collapse
Affiliation(s)
- Jenny Sassone
- San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - GiuliaMaia Serratto
- CNR Institute of Neuroscience, Department BIOMETRA, Università degli Studi di Milano, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - Vincenzo Silani
- IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy.,Department of Pathophysiology and Transplantation, 'Dino Ferrari' Centre, Università degli Studi di Milano, Milan, Italy
| | - Maria Passafaro
- CNR Institute of Neuroscience, Department BIOMETRA, Università degli Studi di Milano, Milan, Italy
| | - Andrea Ciammola
- IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy
| |
Collapse
|
20
|
F Hernández L, Castela I, Ruiz-DeDiego I, Obeso JA, Moratalla R. Striatal activation by optogenetics induces dyskinesias in the 6-hydroxydopamine rat model of Parkinson disease. Mov Disord 2017; 32:530-537. [PMID: 28256089 DOI: 10.1002/mds.26947] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/11/2017] [Accepted: 01/16/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Long-term levodopa (l-dopa) treatment is associated with the development of l-dopa-induced dyskinesias in the majority of patients with Parkinson disease (PD). The etiopathogonesis and mechanisms underlying l-dopa-induced dyskinesias are not well understood. METHODS We used striatal optogenetic stimulation to induce dyskinesias in a hemiparkinsonian model of PD in rats. Striatal dopamine depletion was induced unilaterally by 6-hydroxydopamine injection into the medial forebrain bundle. For the optogenetic manipulation, we injected adeno-associated virus particles expressing channelrhodopsin to stimulate striatal medium spiny neurons with a laser source. RESULTS Simultaneous optical activation of medium spiny neurons of the direct and indirect striatal pathways in the 6-hydroxydopamine lesion but l-dopa naïve rats induced involuntary movements similar to l-dopa-induced dyskinesias, labeled here as optodyskinesias. Noticeably, optodyskinesias were facilitated by l-dopa in animals that did not respond initially to the laser stimulation. In general, optodyskinesias lasted while the laser stimulus was applied, but in some instances remained ongoing for a few seconds after the laser was off. Postmortem tissue analysis revealed increased FosB expression, a molecular marker of l-dopa-induced dyskinesias, primarily in medium spiny neurons of the direct pathway in the dopamine-depleted hemisphere. CONCLUSION Selective optogenetic activation of the dorsolateral striatum elicits dyskinesias in the 6-hydroxydopamine rat model of PD. This effect was associated with a preferential activation of the direct striato-nigral pathway. These results potentially open new avenues in the understanding of mechanisms involved in l-dopa-induced dyskinesias. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ledia F Hernández
- HM-CINAC, Hospital Universitario HM Puerta del Sur, Mostoles and Medical School, CEU-San Pablo University, Madrid, Spain.,CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Ivan Castela
- HM-CINAC, Hospital Universitario HM Puerta del Sur, Mostoles and Medical School, CEU-San Pablo University, Madrid, Spain
| | - Irene Ruiz-DeDiego
- CIBERNED, Instituto Carlos III, Madrid, Spain.,Instituto Cajal-CSIC, Madrid, Spain
| | - Jose A Obeso
- HM-CINAC, Hospital Universitario HM Puerta del Sur, Mostoles and Medical School, CEU-San Pablo University, Madrid, Spain.,CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Rosario Moratalla
- CIBERNED, Instituto Carlos III, Madrid, Spain.,Instituto Cajal-CSIC, Madrid, Spain
| |
Collapse
|
21
|
Rapamycin upregulates glutamate transporter and IL-6 expression in astrocytes in a mouse model of Parkinson's disease. Cell Death Dis 2017; 8:e2611. [PMID: 28182002 PMCID: PMC5386462 DOI: 10.1038/cddis.2016.491] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/22/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023]
Abstract
Rapamycin protects mice against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced loss of dopaminergic neurons, which is an established model for Parkinson's disease. We demonstrated that rapamycin preserves astrocytic expression of glutamate transporters and glutamate reuptake. The protective effect was also observed in astrocyte cultures, indicating that rapamycin acts directly on astrocytes. In the MPTP model, rapamycin caused reduced expression of the E3 ubiquitin ligase Nedd4-2 (neuronal precursor cell expressed developmentally downregulated 4-2) and reduced colocalization of glutamate transporters with ubiquitin. Rapamycin increased interleukin-6 (IL-6) expression, which was associated with reduced expression of inflammatory cytokines, indicating anti-inflammatory properties of IL-6 in the MPTP model. NF-κB was shown to be a key mediator for rapamycin, whereas Janus kinase 2, signal transducer and activator of transcription 3, phosphoinositide 3-kinase, and Akt partially mediated rapamycin effects in astrocytes. These results demonstrate for the first time in a Parkinson's disease animal model that the neuroprotective effects of rapamycin are associated with glial and anti-inflammatory effects.
Collapse
|
22
|
Zhang M, Mu H, Shang Z, Kang K, Lv H, Duan L, Li J, Chen X, Teng Y, Jiang Y, Zhang R. Genome-wide pathway-based association analysis identifies risk pathways associated with Parkinson's disease. Neuroscience 2016; 340:398-410. [PMID: 27840232 DOI: 10.1016/j.neuroscience.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. It is generally believed that it is influenced by both genetic and environmental factors, but the precise pathogenesis of PD is unknown to date. In this study, we performed a pathway analysis based on genome-wide association study (GWAS) to detect risk pathways of PD in three GWAS datasets. We first mapped all SNP markers to autosomal genes in each GWAS dataset. Then, we evaluated gene risk values using the minimum P-value of the tagSNPs. We took a pathway as a unit to identify the risk pathways based on the cumulative risks of the genes in the pathway. Finally, we combine the analysis results of the three datasets to detect the high risk pathways associated with PD. We found there were five same pathways in the three datasets. Besides, we also found there were five pathways which were shared in two datasets. Most of these pathways are associated with nervoussystem. Five pathways had been reported to be PD-related pathways in the previous literature. Our findings also implied that there was a close association between immune response and PD. Continued investigation of these pathways will further help us explain the pathogenesis of PD.
Collapse
Affiliation(s)
- Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongbo Mu
- College of Science, Northeast Forestry University, Harbin, China
| | - Zhenwei Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Kang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lian Duan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinren Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanbo Teng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
23
|
Transcriptome Profile Changes in Mice with MPTP-Induced Early Stages of Parkinson's Disease. Mol Neurobiol 2016; 54:6775-6784. [PMID: 27757834 DOI: 10.1007/s12035-016-0190-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/30/2016] [Indexed: 01/26/2023]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Despite progress in the study of the molecular, genetic, and pathogenic mechanisms of PD, it is unclear which processes trigger the development of the pathology associated with PD. Models of the presymptomatic and early symptomatic stages of PD induced by MPTP have been used to analyze changes in transcriptome profile in brain tissues, to identify specific patterns and mechanisms underlying neurodegeneration in PD. The whole-transcriptome analysis in the brain tissues of the mice with MPTP-induced PD showed that striatum is involved in the pathogenesis in the earliest stages and the processes associated with vesicular transport may be altered. The expression profiles of the genes studied in the substantia nigra and peripheral blood confirm that lymphocytes from peripheral blood may reflect processes occurring in the brain. These data suggest that messenger RNA (mRNA) levels in peripheral blood may provide potential biomarkers of the neurodegeneration occurring in PD. The changes in expression at the mRNA and protein levels suggest that Snca may be involved in neurodegeneration and Drd2 may participate in the development of the compensatory mechanisms in the early stages of PD pathogenesis. Our data suggest that the brain cortex may be involved in the pathological processes in the early stages of PD, including the presymptomatic stage.
Collapse
|
24
|
Rauti R, Lozano N, León V, Scaini D, Musto M, Rago I, Ulloa Severino FP, Fabbro A, Casalis L, Vázquez E, Kostarelos K, Prato M, Ballerini L. Graphene Oxide Nanosheets Reshape Synaptic Function in Cultured Brain Networks. ACS NANO 2016; 10:4459-71. [PMID: 27030936 DOI: 10.1021/acsnano.6b00130] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene offers promising advantages for biomedical applications. However, adoption of graphene technology in biomedicine also poses important challenges in terms of understanding cell responses, cellular uptake, or the intracellular fate of soluble graphene derivatives. In the biological microenvironment, graphene nanosheets might interact with exposed cellular and subcellular structures, resulting in unexpected regulation of sophisticated biological signaling. More broadly, biomedical devices based on the design of these 2D planar nanostructures for interventions in the central nervous system require an accurate understanding of their interactions with the neuronal milieu. Here, we describe the ability of graphene oxide nanosheets to down-regulate neuronal signaling without affecting cell viability.
Collapse
Affiliation(s)
- Rossana Rauti
- Life Science Department, University of Trieste , 34127 Trieste, Italy
| | - Neus Lozano
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Veronica León
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Denis Scaini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | - Mattia Musto
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| | - Ilaria Rago
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | | | - Alessandra Fabbro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
| | | | - Ester Vázquez
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
- CIC BiomaGUNE, Parque Tecnológico de San Sebastián, Paseo Miramón, 182, 20009 San Sebastián, Guipúzcoa, Spain
- Basque Foundation for Science , Ikerbasque, Bilbao 48013, Spain
| | - Laura Ballerini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| |
Collapse
|
25
|
Recent Advance in the Relationship between Excitatory Amino Acid Transporters and Parkinson's Disease. Neural Plast 2016; 2016:8941327. [PMID: 26981287 PMCID: PMC4769779 DOI: 10.1155/2016/8941327] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/05/2016] [Accepted: 01/18/2016] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease (PD) is the most common movement disorder disease in the elderly and is characterized by degeneration of dopamine neurons and formation of Lewy bodies. Glutamate is the major excitatory neurotransmitter in the central nervous system (CNS). If glutamate is not removed promptly in the synaptic cleft, it will excessively stimulate the glutamate receptors and induce excitotoxic effects on the CNS. With lack of extracellular enzyme to decompose glutamate, glutamate uptake in the synaptic cleft is mainly achieved by the excitatory amino acid transporters (EAATs, also known as high-affinity glutamate transporters). Current studies have confirmed that decreased expression and function of EAATs appear in PD animal models. Moreover, single unilateral administration of EAATs inhibitor in the substantia nigra mimics several PD features and this is a solid evidence supporting that decreased EAATs contribute to the process of PD. Drugs or treatments promoting the expression and function of EAATs are shown to attenuate dopamine neurons death in the substantia nigra and striatum, ameliorate the behavior disorder, and improve cognitive abilities in PD animal models. EAATs are potential effective drug targets in treatment of PD and thus study of relationship between EAATs and PD has predominant medical significance currently.
Collapse
|
26
|
Takahashi K, Foster JB, Lin CLG. Glutamate transporter EAAT2: regulation, function, and potential as a therapeutic target for neurological and psychiatric disease. Cell Mol Life Sci 2015; 72:3489-506. [PMID: 26033496 PMCID: PMC11113985 DOI: 10.1007/s00018-015-1937-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the central nervous system. Excitatory amino acid transporter 2 (EAAT2) is primarily responsible for clearance of extracellular glutamate to prevent neuronal excitotoxicity and hyperexcitability. EAAT2 plays a critical role in regulation of synaptic activity and plasticity. In addition, EAAT2 has been implicated in the pathogenesis of many central nervous system disorders. In this review, we summarize current understanding of EAAT2, including structure, pharmacology, physiology, and functions, as well as disease relevancy, such as in stroke, Parkinson's disease, epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, major depressive disorder, and addiction. A large number of studies have demonstrated that up-regulation of EAAT2 protein provides significant beneficial effects in many disease models suggesting EAAT2 activation is a promising therapeutic approach. Several EAAT2 activators have been identified. Further understanding of EAAT2 regulatory mechanisms could improve development of drug-like compounds that spatiotemporally regulate EAAT2.
Collapse
Affiliation(s)
- Kou Takahashi
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Joshua B. Foster
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|