1
|
Tao M, Zhang LL, Zhou GH, Wang C, Luo X. Inhibition of metabotropic glutamate receptor-5 alleviates hepatic steatosis by enhancing autophagy via activation of the AMPK signaling pathway. World J Gastroenterol 2025; 31:98852. [DOI: 10.3748/wjg.v31.i7.98852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/08/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has continued to increase annually. Recent studies have indicated that inhibition of metabotropic glutamate receptor 5 (mGluR5) may alleviate hepatic steatosis. However, the precise mechanism warrants further exploration.
AIM To investigate the potential mechanism by which mGluR5 attenuates hepatocyte steatosis in vitro and in vivo.
METHODS Free fatty acids (FFAs)-stimulated HepG2 cells were treated with the mGluR5 antagonist MPEP and the mGluR5 agonist CHPG. Oil Red O staining and a triglyceride assay kit were used to evaluate lipid content. Western blot analysis was conducted to detect the expression of the autophagy-associated proteins p62 and LC3-II, as well as the expression of the key signaling molecules AMPK and ULK1, in the treated cells. To further elucidate the contributions of autophagy and AMPK, we used chloroquine (CQ) to inhibit autophagy and compound C (CC) to inhibit AMPK activity. In parallel, wild-type mice and mGluR5 knockout (KO) mice fed a normal chow diet or a high-fat diet (HFD) were used to evaluate the effect of mGluR5 inhibition in vivo.
RESULTS mGluR5 inhibition by MPEP attenuated hepatocellular steatosis and increased LC3-II and p62 protein expression. The autophagy inhibitor CQ reversed the effects of MPEP. In addition, MPEP promoted AMPK and ULK1 expression in HepG2 cells exposed to FFAs. MPEP treatment led to the nuclear translocation of transcription factor EB, which is known to promote p62 expression. This effect was negated by the AMPK inhibitor CC. mGluR5 KO mice presented reduced body weight, improved glucose tolerance and reduced hyperlipidemia when fed a HFD. Additionally, the livers of HFD-fed mGluR5 KO mice presented increases in LC3-II and p62.
CONCLUSION Our results suggest that mGluR5 inhibition promoted autophagy and reduced hepatocyte steatosis through activation of the AMPK signaling pathway. These findings reveal a new functional mechanism of mGluR5 as a target in the treatment of MASLD.
Collapse
Affiliation(s)
- Min Tao
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Li-Li Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guang-Hong Zhou
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Cong Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xie Luo
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Orzeł U, Barreto CAV, Filipek S, Moreira IS. GPCR oligomerization across classes: A2AR-mediated regulation of mGlu5R activation. Int J Biol Macromol 2025; 299:139880. [PMID: 39842585 DOI: 10.1016/j.ijbiomac.2025.139880] [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: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
The adenosine A2A receptor (A2AR), a class A GPCR, is a known player in neurological diseases, including Parkinson's disease and Alzheimer's disease, and is also implicated in SARS-CoV-2 infection. Recent studies have revealed its oligomerization with metabotropic glutamate receptor type 5 (mGlu5R), a class C G protein coupled receptor (GPCR) that exists in the homodimeric form. Simultaneous activation of both receptors synergistically enhances mGlu5R-mediated effects in the hippocampus. Despite their importance, the molecular mechanisms governing these interactions remain unclear. In this study, we used molecular modelling techniques, including molecular docking, extensive molecular dynamics (MD) simulations, and detailed analysis, to elucidate the interactions between mGlu5R and A2AR in the inactive and active states. Our findings provide molecular-level insights into the permissive role of A2AR in mGlu5R activation, demonstrating that the inactive A2AR interface within the oligomer blocks the mGlu5R transmembrane helix 6 (TM6), which is crucial for activation. Upon A2AR activation, the oligomer interface undergoes conformational rearrangement, exposing mGlu5R-TM6 and allowing for mGlu5R activation. Furthermore, we identified a pivotal role of the mGlu5R-TM4:A2AR-TM4 interface in facilitating mGlu5R activation. These results highlight the intricate architecture of the mGlu5R:A2AR oligomer, advancing our understanding of GPCR oligomerization and its regulatory mechanisms on receptor activity.
Collapse
Affiliation(s)
- Urszula Orzeł
- PhD Programme in Biosciences, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland; Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Carlos A V Barreto
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Sławomir Filipek
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland; Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| |
Collapse
|
3
|
Puranik N, Song M. Glutamate: Molecular Mechanisms and Signaling Pathway in Alzheimer's Disease, a Potential Therapeutic Target. Molecules 2024; 29:5744. [PMID: 39683904 DOI: 10.3390/molecules29235744] [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: 11/08/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Gamma-glutamate is an important excitatory neurotransmitter in the central nervous system (CNS), which plays an important role in transmitting synapses, plasticity, and other brain activities. Nevertheless, alterations in the glutamatergic signaling pathway are now accepted as a central element in Alzheimer's disease (AD) pathophysiology. One of the most prevalent types of dementia in older adults is AD, a progressive neurodegenerative illness brought on by a persistent decline in cognitive function. Since AD has been shown to be multifactorial, a variety of pharmaceutical targets may be used to treat the condition. N-methyl-D-aspartic acid receptor (NMDAR) antagonists and acetylcholinesterase inhibitors (AChEIs) are two drug classes that the Food and Drug Administration has authorized for the treatment of AD. The AChEIs approved to treat AD are galantamine, donepezil, and rivastigmine. However, memantine is the only non-competitive NMDAR antagonist that has been authorized for the treatment of AD. This review aims to outline the involvement of glutamate (GLU) at the molecular level and the signaling pathways that are associated with AD to demonstrate the drug target therapeutic potential of glutamate and its receptor. We will also consider the opinion of the leading authorities working in this area, the drawback of the existing therapeutic strategies, and the direction for the further investigation.
Collapse
Affiliation(s)
- Nidhi Puranik
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
4
|
Zou ZZ, Han MJ, Chang Y, Li G. Insights into the Metabolism, Disposition, and Quantitative Profile of mGlu5 NAM AE90015 with Metabolite Identification and a Novel Integration Method. Molecules 2024; 29:5724. [PMID: 39683882 DOI: 10.3390/molecules29235724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
AE90015 is a highly specific and effective negative allosteric modulator (NAM) for the human mGlu5 receptor, showing significant promise for treating Parkinson's disease. An in vivo rat oral dose study was conducted on AE90015, which involved the collection of urine and bile samples over a 24 h period. At the study's endpoint, plasma, liver, brain, and renal tissues were also collected. A total of 30 metabolites of AE90015 were identified and structurally characterized or detected using high-resolution LC-MS/MSn. These metabolites fall into four categories: mono-hydroxyl, di-hydroxyl, mono-hydroxyl glucuronide, and di-hydroxyl glucuronide. This study provided a comprehensive overview of the metabolism, excretion, and disposition of AE90015, a promising NAM. The primary clearance pathway for AE90015 is mono-oxidation, accounting for 96% of the total, while direct excretion via renal and bile routes accounted for only 0.5%. Bile emerged as the predominant excretion route, at 65%, for metabolites and a minor amount of parent compound, which contrasts with the common assumption that urine would be the primary excretion pathway, which accounted for 26%. Each adamantyl and pyrazine moiety of AE90015 undergoes a one-time oxidation, while the pyridyl portion remains unmetabolized. Secondary metabolites, such as di-hydroxylated forms and glucuronide conjugates, do not contribute to clearance. In this work, a new quantification method combining UV and mass spectra integration was developed, allowing for the quantification of overlapping metabolite peaks. This novel approach proved to be highly effective for metabolite identification in early preclinical studies.
Collapse
Affiliation(s)
- Zhiyang Zack Zou
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Ming-Jie Han
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Yu Chang
- Department of DMPK & TOX, Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, Beijing 100192, China
| | - Guiying Li
- TB Alliance, 80 Pine St. 20th Floor, New York, NY 10005, USA
| |
Collapse
|
5
|
Tang R, Franz CE, Hauger RL, Dale AM, Dorros SM, Eyler LT, Fennema-Notestine C, Hagler DJ, Lyons MJ, Panizzon MS, Puckett OK, Williams ME, Elman JA, Kremen WS. Early Cortical Microstructural Changes in Aging Are Linked to Vulnerability to Alzheimer's Disease Pathology. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:975-985. [PMID: 38878863 PMCID: PMC11756816 DOI: 10.1016/j.bpsc.2024.05.012] [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: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Early identification of Alzheimer's disease (AD) risk is critical for improving treatment success. Cortical thickness is a macrostructural measure used to assess neurodegeneration in AD. However, cortical microstructural changes appear to precede macrostructural atrophy and may improve early risk identification. Currently, whether cortical microstructural changes in aging are linked to vulnerability to AD pathophysiology remains unclear in nonclinical populations, who are precisely the target for early risk identification. METHODS In 194 adults, we calculated magnetic resonance imaging-derived maps of changes in cortical mean diffusivity (microstructure) and cortical thickness (macrostructure) over 5 to 6 years (mean age: time 1 = 61.82 years; time 2 = 67.48 years). Episodic memory was assessed using 3 well-established tests. We obtained positron emission tomography-derived maps of AD pathology deposition (amyloid-β, tau) and neurotransmitter receptors (cholinergic, glutamatergic) implicated in AD pathophysiology. Spatial correlational analyses were used to compare pattern similarity among maps. RESULTS Spatial patterns of cortical macrostructural changes resembled patterns of cortical organization sensitive to age-related processes (r = -0.31, p < .05), whereas microstructural changes resembled the patterns of tau deposition in AD (r = 0.39, p = .038). Individuals with patterns of microstructural changes that more closely resembled stereotypical tau deposition exhibited greater memory decline (β = 0.22, p = .029). Microstructural changes and AD pathology deposition were enriched in areas with greater densities of cholinergic and glutamatergic receptors (ps < .05). CONCLUSIONS Patterns of cortical microstructural changes were more AD-like than patterns of macrostructural changes, which appeared to reflect more general aging processes. Microstructural changes may better inform early risk prediction efforts as a sensitive measure of vulnerability to pathological processes prior to overt atrophy and cognitive decline.
Collapse
Affiliation(s)
- Rongxiang Tang
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California.
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - Richard L Hauger
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California; Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Stephen M Dorros
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, California; Desert Pacific Mental Illness Research Education and Clinical Center, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California; Department of Radiology, University of California San Diego, La Jolla, California
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - McKenna E Williams
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California; Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California San Diego, San Diego, California
| | - Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| |
Collapse
|
6
|
Wang J, Huang Q, He K, Li J, Guo T, Yang Y, Lin Z, Li S, Vanderlinden G, Huang Y, Van Laere K, Guan Y, Guo Q, Ni R, Li B, Xie F. Presynaptic density determined by SV2A PET is closely associated with postsynaptic metabotropic glutamate receptor 5 availability and independent of amyloid pathology in early cognitive impairment. Alzheimers Dement 2024; 20:3876-3888. [PMID: 38634334 PMCID: PMC11180932 DOI: 10.1002/alz.13817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Metabotropic glutamate receptor 5 (mGluR5) is involved in regulating integrative brain function and synaptic transmission. Aberrant mGluR5 signaling and relevant synaptic failure play a key role in the pathophysiological mechanism of Alzheimer's disease (AD). METHODS Ten cognitively impaired (CI) individuals and 10 healthy controls (HCs) underwent [18F]SynVesT-1 and [18F]PSS232 positron emission tomography (PET)/magnetic resonance to assess synaptic density and mGluR5 availability. The associations between mGluR5 availability and synaptic density were examined. A mediation analysis was performed to investigate the possible mediating effects of mGluR5 availability and synaptic loss on the relationship between amyloid deposition and cognition. RESULTS CI patients exhibited lower mGluR5 availability and synaptic density in the medial temporal lobe than HCs. Regional synaptic density was closely associated with regional mGluR5 availability. mGluR5 availability and synaptic loss partially mediated the relationship between amyloid deposition and cognition. CONCLUSIONS Reductions in mGluR5 availability and synaptic density exhibit similar spatial patterns in AD and are closely linked. HIGHLIGHTS Cognitively impaired patients exhibited lower mGluR5 availability and synaptic density in the medial temporal lobe than HCs. Reductions in mGluR5 availability and synaptic density exhibit similar spatial patterns in AD. Regional synaptic density was closely associated with regional mGluR5 availability. mGluR5 availability and synaptic loss partially mediated the relationship between amyloid deposition and global cognition. With further research, modulating mGluR5 availability might be a potential therapeutic strategy for improving synaptic function in AD.
Collapse
Affiliation(s)
- Jie Wang
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qi Huang
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Kun He
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Junpeng Li
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay LaboratoryShenzhenChina
| | - Yang Yang
- Beijing United Imaging Research Institute of Intelligent ImagingBeijingChina
| | - Zengping Lin
- Central Research Institute, United Imaging Healthcare Group Co., LtdShanghaiChina
| | - Songye Li
- Department of Radiology and Biomedical ImagingPET CenterYale University School of MedicineNew HavenConnecticutUSA
| | - Greet Vanderlinden
- Department of Imaging and PathologyNuclear Medicine and Molecular Imaging, KU LeuvenLeuvenBelgium
| | - Yiyun Huang
- Department of Radiology and Biomedical ImagingPET CenterYale University School of MedicineNew HavenConnecticutUSA
| | - Koen Van Laere
- Department of Imaging and PathologyNuclear Medicine and Molecular Imaging, KU LeuvenLeuvenBelgium
| | - Yihui Guan
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qihao Guo
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich & ETH ZurichZurichSwitzerland
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Binying Li
- Department of Neurology and Institute of NeurologyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Xie
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
7
|
Fotuhi SN, Khalaj-Kondori M. Imbalanced clearance of Aβ peptide cause presynaptic plaque formation. Int J Neurosci 2024; 134:66-70. [PMID: 35639020 DOI: 10.1080/00207454.2022.2085099] [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: 08/03/2020] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease is characterized by abnormal increase of Aβ peptide which is likely as the result of imbalanced homeostasis of its production and clearance mechanisms. Here, we briefly review that the uncleaned extracellular Aβ peptides are loaded into presynaptic neurons. The Aβ oligomers desperately affect pre- and post-synapse neuron activity and turn into plaques inside the presynaptic neurons over the time passes.
Collapse
Affiliation(s)
- Seyedeh Nahid Fotuhi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
8
|
Wang Y, Huang Y, Ma A, You J, Miao J, Li J. Natural Antioxidants: An Effective Strategy for the Treatment of Alzheimer's Disease at the Early Stage. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:11854-11870. [PMID: 38743017 DOI: 10.1021/acs.jafc.4c01323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The critical role of oxidative stress in Alzheimer's disease (AD) has been recognized by researchers recently, and natural antioxidants have been demonstrated to have anti-AD activity in animal models, such as Ginkgo biloba extract, soy isoflavones, lycopene, and so on. This paper summarized these natural antioxidants and points out that natural antioxidants always have multiple advantages which are help to deal with AD, such as clearing free radicals, regulating signal transduction, protecting mitochondrial function, and synaptic plasticity. Based on the available data, we have created a relatively complete pathway map of reactive oxygen species (ROS) and AD-related targets and concluded that oxidative stress caused by ROS is the core of AD pathogenesis. In the prospect, we introduced the concept of a combined therapeutic strategy, termed "Antioxidant-Promoting Synaptic Remodeling," highlighting the integration of antioxidant interventions with synaptic remodeling approaches as a novel avenue for therapeutic exploration.
Collapse
Affiliation(s)
- Yifeng Wang
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| | - Yan Huang
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| | - Aixia Ma
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| | - Jiahe You
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| | - Jing Miao
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi, Xinjiang 830000, PR China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi, Xinjiang 830000, PR China
- National Demonstration Center for Experimental Biology Education, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| | - Jinyao Li
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830000, PR China
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi, Xinjiang 830000, PR China
- National Demonstration Center for Experimental Biology Education, Xinjiang University, Urumqi, Xinjiang 830000, PR China
| |
Collapse
|
9
|
Aghamiri H, Jafari-Sabet M, Hoormand M. Ameliorative Effect of Cannabidiol on Topiramate-Induced Memory Loss: The Role of Hippocampal and Prefrontal Cortical NMDA Receptors and CREB/BDNF Signaling Pathways in Rats. Neurochem Res 2024; 49:363-378. [PMID: 37814133 DOI: 10.1007/s11064-023-04041-4] [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: 07/27/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Cannabidiol (CBD) is a promising neurological agent with potential beneficial effects on memory and cognitive function. The combination of CBD and topiramate in the treatment of some neurological diseases has been of great interest. Since Topiramate-induced memory loss is a major drawback of its clinical application and the overall effect of the combination of CBD and topiramate on memory is still unclear, here we investigated the effect of CBD on topiramate-induced memory loss and the underlying molecular mechanisms. A one trial step-through inhibitory test was used to evaluate memory consolidation in rats. Moreover, the role of N-methyl-D-aspartate receptors (NMDARs) in the combination of CBD and topiramate in memory consolidation was evaluated through the intra-CA1 administration of MK-801 and NMDA. Western blot analysis was used to evaluate variations in brain-derived neurotrophic factor (BDNF) and phosphorylated cyclic AMP response element-binding protein (pCREB)/CREB ratio in the prefrontal cortex (PFC) and hippocampus (HPC). While the intraperitoneal (i.p.) administration of topiramate (50, 75, and 100 mg/kg) significantly reduced inhibitory time latency, the i.p. administration of CBD (20 and 40 mg/kg) could effectively reverse these effects. Similarly, the sub-effective doses of NMDA plus CBD (10 mg/kg) could improve the topiramate-induced memory loss along with an enhancement in BDNF and pCREB expression in the PFC and HPC. Contrarily, the administration of sub-effective doses of the NMDAR antagonist (MK-801) diminished the protective effects of CBD (20 mg/kg) on topiramate-induced memory loss associated with decreased BDNF and pCREB levels in the PFC and HPC. These findings suggest that CBD can improve topiramate-induced memory impairment, partially by the NMDARs of the PFC and HPC, possibly regulated by the CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Helia Aghamiri
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Majid Jafari-Sabet
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahmood Hoormand
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Love RWB. Aniracetam: An Evidence-Based Model for Preventing the Accumulation of Amyloid-β Plaques in Alzheimer's Disease. J Alzheimers Dis 2024; 98:1235-1241. [PMID: 38552113 DOI: 10.3233/jad-231247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Alzheimer's disease is the leading cause of dementia in the world. It affects 6 million people in the United States and 50 million people worldwide. Alzheimer's disease is characterized by the accumulation of amyloid-β plaques (Aβ), an increase in tau protein neurofibrillary tangles, and a loss of synapses. Since the 1990s, removing and reducing Aβ has been the focus of Alzheimer's treatment and prevention research. The accumulation of Aβ can lead to oxidative stress, inflammation, neurotoxicity, and eventually apoptosis. These insults impair signaling systems in the brain, potentially leading to memory loss and cognitive decline. Aniracetam is a safe, effective, cognitive-enhancing drug that improves memory in both human and animal studies. Aniracetam may prevent the production and accumulation of Aβ by increasing α-secretase activity through two distinct pathways: 1) increasing brain derived neurotrophic factor expression and 2) positively modulating metabotropic glutamate receptors. This is the first paper to propose an evidence-based model for aniracetam reducing the accumulation and production of Aβ.
Collapse
Affiliation(s)
- Robert W B Love
- Research Department, Brain Fit For Life, LLC, Lewes, DE, USA
| |
Collapse
|
11
|
Gautam D, Naik UP, Naik MU, Yadav SK, Chaurasia RN, Dash D. Glutamate Receptor Dysregulation and Platelet Glutamate Dynamics in Alzheimer's and Parkinson's Diseases: Insights into Current Medications. Biomolecules 2023; 13:1609. [PMID: 38002291 PMCID: PMC10669830 DOI: 10.3390/biom13111609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Two of the most prevalent neurodegenerative disorders (NDDs), Alzheimer's disease (AD) and Parkinson's disease (PD), present significant challenges to healthcare systems worldwide. While the etiologies of AD and PD differ, both diseases share commonalities in synaptic dysfunction, thereby focusing attention on the role of neurotransmitters. The possible functions that platelets may play in neurodegenerative illnesses including PD and AD are becoming more acknowledged. In AD, platelets have been investigated for their ability to generate amyloid-ß (Aß) peptides, contributing to the formation of neurotoxic plaques. Moreover, platelets are considered biomarkers for early AD diagnosis. In PD, platelets have been studied for their involvement in oxidative stress and mitochondrial dysfunction, which are key factors in the disease's pathogenesis. Emerging research shows that platelets, which release glutamate upon activation, also play a role in these disorders. Decreased glutamate uptake in platelets has been observed in Alzheimer's and Parkinson's patients, pointing to a systemic dysfunction in glutamate handling. This paper aims to elucidate the critical role that glutamate receptors play in the pathophysiology of both AD and PD. Utilizing data from clinical trials, animal models, and cellular studies, we reviewed how glutamate receptors dysfunction contributes to neurodegenerative (ND) processes such as excitotoxicity, synaptic loss, and cognitive impairment. The paper also reviews all current medications including glutamate receptor antagonists for AD and PD, highlighting their mode of action and limitations. A deeper understanding of glutamate receptor involvement including its systemic regulation by platelets could open new avenues for more effective treatments, potentially slowing disease progression and improving patient outcomes.
Collapse
Affiliation(s)
- Deepa Gautam
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Ulhas P. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Meghna U. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Santosh K. Yadav
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Rameshwar Nath Chaurasia
- The Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India;
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
12
|
Kim Y, Kim J, Kang S, Chang KA. Depressive-like Behaviors Induced by mGluR5 Reduction in 6xTg in Mouse Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:13010. [PMID: 37629191 PMCID: PMC10455602 DOI: 10.3390/ijms241613010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD) is one representative dementia characterized by the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain, resulting in cognitive decline and memory loss. AD is associated with neuropsychiatric symptoms, including major depressive disorder (MDD). Recent studies showed a reduction in mGluR5 expression in the brains of stress-induced mice models and individuals with MDD compared to controls. In our study, we identified depressive-like behavior and memory impairment in a mouse model of AD, specifically in the 6xTg model with tau and Aβ pathologies. In addition, we investigated the expression of mGluR5 in the brains of 6xTg mice using micro-positron emission tomography (micro-PET) imaging, histological analysis, and Western blot analysis, and we observed a decrease in mGluR5 levels in the brains of 6xTg mice compared to wild-type (WT) mice. Additionally, we identified alterations in the ERK/AKT/GSK-3β signaling pathway in the brains of 6xTg mice. Notably, we identified a significant negative correlation between depressive-like behavior and the protein level of mGluR5 in 6xTg mice. Additionally, we also found a significant positive correlation between depressive-like behavior and AD pathologies, including phosphorylated tau and Aβ. These findings suggested that abnormal mGluR5 expression and AD-related pathologies were involved in depressive-like behavior in the 6xTg mouse model. Further research is warranted to elucidate the underlying mechanisms and explore potential therapeutic targets in the intersection of AD and depressive-like symptoms.
Collapse
Affiliation(s)
- Youngkyo Kim
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
| | - Jinho Kim
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, VT 55905, USA
| | - Keun-A Chang
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea
| |
Collapse
|
13
|
Holter KM, Pierce BE, Gould RW. Metabotropic glutamate receptor function and regulation of sleep-wake cycles. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:93-175. [PMID: 36868636 PMCID: PMC10973983 DOI: 10.1016/bs.irn.2022.11.002] [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] [Indexed: 01/15/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer's disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bethany E Pierce
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Robert W Gould
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
14
|
Role of amyloid beta (25-35) neurotoxicity in the ferroptosis and necroptosis as modalities of regulated cell death in Alzheimer's disease. Neurotoxicology 2023; 94:71-86. [PMID: 36347329 DOI: 10.1016/j.neuro.2022.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Neuronal cell death as a prominent pathological feature contributes to cognitive decline and memory loss in Alzheimer's disease. We investigated the role of two forms of cell death pathways, ferroptosis and necroptosis, and their interactions following entorhinal cortex (EC) amyloidopathy. The Aβ25-35 was bilaterally injected into the rat's EC, and Morris Water Maze was applied to determine spatial performance one week after Aβ injection. For evaluation of ferroptosis and necroptosis involvement in Aβ induced pathology, ferroptosis inhibitor, Ferrostatin (Fer-1), and necroptosis inhibitor, Necrostatin (Nec-1), were injected into the EC during training days of behavioral test. Our behavioral and histological assessment showed spatial learning and memory impairment, along with neuropathology changes such as cell survival and intracellular Aβ deposits in response to EC amyloidopathy, which were ameliorated by treatment with Fer-1 or Nec-1. The expression of ferroptosis key factors GPX4 and SLC7A11 were decreased and the level of TfR was increased following Aβ toxicity. Also, Necroptosis pathway related factors RIP1, RIP3, and MLKL were modulated by Aβ neurotoxicity. However, application of Fer-1 or Nec-1 could inhibit the hippocampal ferroptosis and necroptosis pathways due to EC amyloidopathy. Our data also demonstrated that Aβ-induced necroptosis suppressed by Fer-1, although Nec-1 had no effect on ferroptosis, indicating that ferroptosis pathway is upstream of necroptosis process in the Aβ neurotoxicity. Moreover, Aβ induced hippocampal mGLUR5 overexpression and reduced level of STIM1/2 recovered by Fer-1 or Nec-1. According to our findings ferroptosis and necroptosis pathways are involved in Aβ neurotoxicity through modulation of mGLUR5 and STIM1/2 signaling.
Collapse
|
15
|
Mazzitelli M, Presto P, Antenucci N, Meltan S, Neugebauer V. Recent Advances in the Modulation of Pain by the Metabotropic Glutamate Receptors. Cells 2022; 11:2608. [PMID: 36010684 PMCID: PMC9406805 DOI: 10.3390/cells11162608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 01/22/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR or mGlu) are G-protein coupled receptors activated by the binding of glutamate, the main classical neurotransmitter of the nervous system. Eight different mGluR subtypes (mGluR1-8) have been cloned and are classified in three groups based on their molecular, pharmacological and signaling properties. mGluRs mediate several physiological functions such as neuronal excitability and synaptic plasticity, but they have also been implicated in numerous pathological conditions including pain. The availability of new and more selective allosteric modulators together with the canonical orthosteric ligands and transgenic technologies has led to significant advances in our knowledge about the role of the specific mGluR subtypes in the pathophysiological mechanisms of various diseases. Although development of successful compounds acting on mGluRs for clinical use has been scarce, the subtype-specific-pharmacological manipulation might be a compelling approach for the treatment of several disorders in humans, including pain; this review aims to summarize and update on preclinical evidence for the roles of different mGluRs in the pain system and discusses knowledge gaps regarding mGluR-related sex differences and neuroimmune signaling in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Shakira Meltan
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
16
|
Mandal AK. Mitochondrial targeting of potent nanoparticulated drugs in combating diseases. J Biomater Appl 2022; 37:614-633. [PMID: 35790487 DOI: 10.1177/08853282221111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mitochondrial dysfunction, characterized by the electron transport chain (ETC) leakage and reduced adenosine tri-phosphate synthesis, occurs primarily due to free radicals -induced mutations in either the mitochondrial deoxyribonucleic acid (mtDNA) or nuclear (n) DNA caused by pathogenic infections, toxicant exposures, adverse drug-effects, or other environmental exposures, leading to secondary dysfunction affecting ischemic, diabetic, cancerous, and degenerative diseases. In these concerns, mitochondria-targeted remedies may include a significant role in the protection and treatment of mitochondrial function to enhance its activity. Coenzyme Q10 pyridinol and pyrimidinol antioxidant analogues and other potent drug-compounds for their multifunctional radical quencher and other anti-toxic activities may take a significant therapeutic effectivity for ameliorating mitochondrial dysfunction. Moreover, the encapsulation of these bioactive ligands-attached potent compounds in vesicular system may enable them a superb biological effective for the treatment of mitochondria-targeted dysfunction-related diseases with least side effects. This review depicts mainly on mitochondrial enzymatic dysfunction and their amelioration by potent drugs with the usages of nanoparticulated delivery system against mitochondria-affected diseases.
Collapse
|
17
|
Hu C, Chen C, Xia Y, Chen J, Yang W, Wang L, Chen DD, Wu YZ, Fan Q, Jia XX, Xiao K, Shi Q, Chen ZB, Dong XP. Different Aberrant Changes of mGluR5 and Its Downstream Signaling Pathways in the Scrapie-Infected Cell Line and the Brains of Scrapie-Infected Experimental Rodents. Front Cell Dev Biol 2022; 10:844378. [PMID: 35646890 PMCID: PMC9133610 DOI: 10.3389/fcell.2022.844378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Metabotropic glutamate receptor subtype 5 (mGluR5) is a G-protein-coupled receptor found widely in the central nervous system. It has been involved in the development and progression of some neurodegenerative diseases, but its role in prion diseases is rarely described. In this study, the changes of mGluR5 and its downstream signaling pathways in prion-infected cell line SMB-S15 and the brains of scrapie-infected experimental rodents were evaluated by various methodologies. We found the levels of mGluR5 were significantly increased in a prion-infected cell line SMB-S15 and the cultured cells transiently express an abnormal form PrP (Cyto-PrP). Using immunoprecipitation tests and immunofluorescent assays (IFA), molecular interaction and morphological colocalization between PrP and mGluR5 were observed in the cultured cells. We identified that the (GPCRs)-IP3-IP3R-Ca2+ pathway was activated and the levels of the downstream kinases p38, ERK, and JNK were increased in SMB-S15 cells. After treated with mGluR5 antagonist (MTEP) or the removal of prion replication by resveratrol in SMB-S15 cells, the upregulations of mGluR5 and the downstream kinases were restored in a certain degree. Moreover, increased mGluR5 contributes to the cell damage in prion-infected cells. Contrarily, the levels of mGluR5 in the brains of several scrapie-infected rodent models were decreased at terminal stage. IFA of the brain sections of scrapie-infected rodents demonstrated that the signals of mGluR5 were preferentially colocalized with the NeuN-positive cells, accompanying with severe neuron losses in Nissl staining, which might be a reason for the decrease of mGluR5. Our data indicate the different aberrant alterations of mGluR5 and the downstream signaling pathways during prion infection in vivo and in vitro.
Collapse
Affiliation(s)
- Chao Hu
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Cao Chen, ; Xiao-Ping Dong,
| | - Ying Xia
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jia Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wei Yang
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lin Wang
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Dong-Dong Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yue-Zhang Wu
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qin Fan
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Xi Jia
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhi-Bao Chen
- College of Agricultural, Guangdong Ocean University, Zhanjiang, China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- China Academy of Chinese Medical Sciences, Beijing, China
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China
- *Correspondence: Cao Chen, ; Xiao-Ping Dong,
| |
Collapse
|
18
|
Meldolesi J. News about Therapies of Alzheimer’s Disease: Extracellular Vesicles from Stem Cells Exhibit Advantages Compared to Other Treatments. Biomedicines 2022; 10:biomedicines10010105. [PMID: 35052785 PMCID: PMC8773509 DOI: 10.3390/biomedicines10010105] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
Upon its discovery, Alzheimer’s, the neurodegenerative disease that affects many millions of patients in the world, remained without an effective therapy. The first drugs, made available near the end of last century, induced some effects, which remained only marginal. More promising effects are now present, induced by two approaches. Blockers of the enzyme BACE-1 induce, in neurons and glial cells, decreased levels of Aβ, the key peptide of the Alzheimer’s disease. If administered at early AD steps, the BACE-1 blockers preclude further development of the disease. However, they have no effect on established, irreversible lesions. The extracellular vesicles secreted by mesenchymal stem cells induce therapy effects analogous, but more convenient, than the effects of their original cells. After their specific fusion to target cells, the action of these vesicles depends on their ensuing release of cargo molecules, such as proteins and many miRNAs, active primarily on the cell cytoplasm. Operationally, these vesicles exhibit numerous advantages: they exclude, by their accurate selection, the heterogeneity of the original cells; exhibit molecular specificity due to their engineering and drug accumulation; and induce effective actions, mediated by variable concentrations of factors and molecules and by activation of signaling cascades. Their strength is reinforced by their combination with various factors and processes. The recent molecular and operations changes, induced especially by the stem cell target cells, result in encouraging and important improvement of the disease. Their further development is expected in the near future.
Collapse
Affiliation(s)
- Jacopo Meldolesi
- San Raffaele Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- Faculty of Medicine, CNR Institute of Neuroscience, University Milano-Bicocca, 20132 Milan, Italy
| |
Collapse
|
19
|
Crestini A, Santilli F, Martellucci S, Carbone E, Sorice M, Piscopo P, Mattei V. Prions and Neurodegenerative Diseases: A Focus on Alzheimer's Disease. J Alzheimers Dis 2021; 85:503-518. [PMID: 34864675 DOI: 10.3233/jad-215171] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Specific protein misfolding and aggregation are mechanisms underlying various neurodegenerative diseases such as prion disease and Alzheimer's disease (AD). The misfolded proteins are involved in prions, amyloid-β (Aβ), tau, and α-synuclein disorders; they share common structural, biological, and biochemical characteristics, as well as similar mechanisms of aggregation and self-propagation. Pathological features of AD include the appearance of plaques consisting of deposition of protein Aβ and neurofibrillary tangles formed by the hyperphosphorylated tau protein. Although it is not clear how protein aggregation leads to AD, we are learning that the cellular prion protein (PrPC) plays an important role in the pathogenesis of AD. Herein, we first examined the pathogenesis of prion and AD with a focus on the contribution of PrPC to the development of AD. We analyzed the mechanisms that lead to the formation of a high affinity bond between Aβ oligomers (AβOs) and PrPC. Also, we studied the role of PrPC as an AβO receptor that initiates an AβO-induced signal cascade involving mGluR5, Fyn, Pyk2, and eEF2K linking Aβ and tau pathologies, resulting in the death of neurons in the central nervous system. Finally, we have described how the PrPC-AβOs interaction can be used as a new potential therapeutic target for the treatment of PrPC-dependent AD.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Santilli
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Stefano Martellucci
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy
| | - Elena Carbone
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| |
Collapse
|
20
|
Abd-Elrahman KS, Ferguson SSG. Noncanonical Metabotropic Glutamate Receptor 5 Signaling in Alzheimer's Disease. Annu Rev Pharmacol Toxicol 2021; 62:235-254. [PMID: 34516293 DOI: 10.1146/annurev-pharmtox-021821-091747] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabotropic glutamate receptor 5 (mGluR5) is ubiquitously expressed in brain regions responsible for memory and learning. It plays a key role in modulating rapid changes in synaptic transmission and plasticity. mGluR5 supports long-term changes in synaptic strength by regulating the transcription and translation of essential synaptic proteins. β-Amyloid 42 (Aβ42) oligomers interact with a mGluR5/cellular prion protein (PrPC) complex to disrupt physiological mGluR5 signal transduction. Aberrant mGluR5 signaling and associated synaptic failure are considered an emerging pathophysiological mechanism of Alzheimer's disease (AD). Therefore, mGluR5 represents an attractive therapeutic target for AD, and recent studies continue to validate the efficacy of various mGluR5 allosteric modulators in improving memory deficits and mitigating disease pathology. However, sex-specific differences in the pharmacology of mGluR5 and activation of noncanonical signaling downstream of the receptor suggest that its utility as a therapeutic target in female AD patients needs to be reconsidered. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Research Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; .,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; email
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Research Institute and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada;
| |
Collapse
|
21
|
Ponroy Bally B, Murai KK. Astrocytes in Down Syndrome Across the Lifespan. Front Cell Neurosci 2021; 15:702685. [PMID: 34483840 PMCID: PMC8416355 DOI: 10.3389/fncel.2021.702685] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/30/2021] [Indexed: 11/23/2022] Open
Abstract
Down Syndrome (DS) is the most common genetic cause of intellectual disability in which delays and impairments in brain development and function lead to neurological and cognitive phenotypes. Traditionally, a neurocentric approach, focusing on neurons and their connectivity, has been applied to understanding the mechanisms involved in DS brain pathophysiology with an emphasis on how triplication of chromosome 21 leads to alterations in neuronal survival and homeostasis, synaptogenesis, brain circuit development, and neurodegeneration. However, recent studies have drawn attention to the role of non-neuronal cells, especially astrocytes, in DS. Astrocytes comprise a large proportion of cells in the central nervous system (CNS) and are critical for brain development, homeostasis, and function. As triplication of chromosome 21 occurs in all cells in DS (with the exception of mosaic DS), a deeper understanding of the impact of trisomy 21 on astrocytes in DS pathophysiology is warranted and will likely be necessary for determining how specific brain alterations and neurological phenotypes emerge and progress in DS. Here, we review the current understanding of the role of astrocytes in DS, and discuss how specific perturbations in this cell type can impact the brain across the lifespan from early brain development to adult stages. Finally, we highlight how targeting, modifying, and/or correcting specific molecular pathways and properties of astrocytes in DS may provide an effective therapeutic direction given the important role of astrocytes in regulating brain development and function.
Collapse
Affiliation(s)
- Blandine Ponroy Bally
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| |
Collapse
|
22
|
Tanaka M, Shigetomi E, Parajuli B, Nagatomo H, Shinozaki Y, Hirayama Y, Saito K, Kubota Y, Danjo Y, Lee JH, Kim SK, Nabekura J, Koizumi S. Adenosine A 2B receptor down-regulates metabotropic glutamate receptor 5 in astrocytes during postnatal development. Glia 2021; 69:2546-2558. [PMID: 34339538 DOI: 10.1002/glia.24006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/07/2022]
Abstract
Metabotropic glutamate receptor 5 (mGluR5) in astrocytes is a key molecule for controlling synapse remodeling. Although mGluR5 is abundant in neonatal astrocytes, its level is gradually down-regulated during development and is almost absent in the adult. However, in several pathological conditions, mGluR5 re-emerges in adult astrocytes and contributes to disease pathogenesis by forming uncontrolled synapses. Thus, controlling mGluR5 expression in astrocyte is critical for several diseases, but the mechanism that regulates mGluR5 expression remains unknown. Here, we show that adenosine triphosphate (ATP)/adenosine-mediated signals down-regulate mGluR5 in astrocytes. First, in situ Ca2+ imaging of astrocytes in acute cerebral slices from post-natal day (P)7-P28 mice showed that Ca2+ responses evoked by (S)-3,5-dihydroxyphenylglycine (DHPG), a mGluR5 agonist, decreased during development, whereas those evoked by ATP or its metabolite, adenosine, increased. Second, ATP and adenosine suppressed expression of the mGluR5 gene, Grm5, in cultured astrocytes. Third, the decrease in the DHPG-evoked Ca2+ responses was associated with down-regulation of Grm5. Interestingly, among several adenosine (P1) receptor and ATP (P2) receptor genes, only the adenosine A2B receptor gene, Adora2b, was up-regulated in the course of development. Indeed, we observed that down-regulation of Grm5 was suppressed in Adora2b knockout astrocytes at P14 and in situ Ca2+ imaging from Adora2b knockout mice indicated that the A2B receptor inhibits mGluR5 expression in astrocytes. Furthermore, deletion of A2B receptor increased the number of excitatory synapse in developmental stage. Taken together, the A2B receptor is critical for down-regulation of mGluR5 in astrocytes, which would contribute to terminate excess synaptogenesis during development.
Collapse
Affiliation(s)
- Masayoshi Tanaka
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Hiroaki Nagatomo
- Center for Life Science Research, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yuri Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yosuke Danjo
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Ji Hwan Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea.,Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
23
|
Castillo CA, Ballesteros-Yáñez I, León-Navarro DA, Albasanz JL, Martín M. Early Effects of the Soluble Amyloid β 25-35 Peptide in Rat Cortical Neurons: Modulation of Signal Transduction Mediated by Adenosine and Group I Metabotropic Glutamate Receptors. Int J Mol Sci 2021; 22:ijms22126577. [PMID: 34205261 PMCID: PMC8234864 DOI: 10.3390/ijms22126577] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/04/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
The amyloid β peptide (Aβ) is a central player in the neuropathology of Alzheimer’s disease (AD). The alteration of Aβ homeostasis may impact the fine-tuning of cell signaling from the very beginning of the disease, when amyloid plaque is not deposited yet. For this reason, primary culture of rat cortical neurons was exposed to Aβ25-35, a non-oligomerizable form of Aβ. Cell viability, metabotropic glutamate receptors (mGluR) and adenosine receptors (AR) expression and signalling were assessed. Aβ25-35 increased mGluR density and affinity, mainly due to a higher gene expression and protein presence of Group I mGluR (mGluR1 and mGluR5) in the membrane of cortical neurons. Intriguingly, the main effector of group I mGluR, the phospholipase C β1 isoform, was less responsive. Also, the inhibitory action of group II and group III mGluR on adenylate cyclase (AC) activity was unaltered or increased, respectively. Interestingly, pre-treatment of cortical neurons with an antagonist of group I mGluR reduced the Aβ25-35-induced cell death. Besides, Aβ25-35 increased the density of A1R and A2AR, along with an increase in their gene expression. However, while A1R-mediated AC inhibition was increased, the A2AR-mediated stimulation of AC remained unchanged. Therefore, one of the early events that takes place after Aβ25-35 exposure is the up-regulation of adenosine A1R, A2AR, and group I mGluR, and the different impacts on their corresponding signaling pathways. These results emphasize the importance of deciphering the early events and the possible involvement of metabotropic glutamate and adenosine receptors in AD physiopathology.
Collapse
Affiliation(s)
- Carlos Alberto Castillo
- Department of Nursing, Physiotherapy and Occupational Therapy, School of Physiotherapy and Nursing, University of Castilla-La Mancha, 45071 Toledo, Spain;
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
| | - Inmaculada Ballesteros-Yáñez
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, School of Medicine of Ciudad Real, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - David Agustín León-Navarro
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, Faculty of Chemical and Technological Sciences, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - José Luis Albasanz
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, School of Medicine of Ciudad Real, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence:
| | - Mairena Martín
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, Faculty of Chemical and Technological Sciences, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| |
Collapse
|
24
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
The Density of Group I mGlu 5 Receptors Is Reduced along the Neuronal Surface of Hippocampal Cells in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22115867. [PMID: 34070808 PMCID: PMC8199018 DOI: 10.3390/ijms22115867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 11/17/2022] Open
Abstract
Metabotropic glutamate receptor subtype 5 (mGlu5) is implicated in the pathophysiology of Alzheimer’s disease (AD). However, its alteration at the subcellular level in neurons is still unexplored. Here, we provide a quantitative description on the expression and localisation patterns of mGlu5 in the APP/PS1 model of AD at 12 months of age, combining immunoblots, histoblots and high-resolution immunoelectron microscopic approaches. Immunoblots revealed that the total amount of mGlu5 protein in the hippocampus, in addition to downstream molecules, i.e., Gq/11 and PLCβ1, was similar in both APP/PS1 mice and age-matched wild type mice. Histoblots revealed that mGlu5 expression in the brain and its laminar expression in the hippocampus was also unaltered. However, the ultrastructural techniques of SDS-FRL and pre-embedding immunogold demonstrated that the subcellular localisation of mGlu5 was significantly reduced along the neuronal surface of hippocampal principal cells, including CA1 pyramidal cells and DG granule cells, in APP/PS1 mice at 12 months of age. The decrease in the surface localisation of mGlu5 was accompanied by an increase in its frequency at intracellular sites in the two neuronal populations. Together, these data demonstrate, for the first time, a loss of mGlu5 at the plasma membrane and accumulation at intracellular sites in different principal cells of the hippocampus in APP/PS1 mice, suggesting an alteration of the excitability and synaptic transmission that could contribute to the cognitive dysfunctions in this AD animal model. Further studies are required to elucidate the specificity of mGlu5-associated molecules and downstream signalling pathways in the progression of the pathology.
Collapse
|
26
|
Identification of functional cytochrome P450 and ferredoxin from Streptomyces sp. EAS-AB2608 by transcriptional analysis and their heterologous expression. Appl Microbiol Biotechnol 2021; 105:4177-4187. [PMID: 33944982 DOI: 10.1007/s00253-021-11304-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/21/2021] [Accepted: 04/18/2021] [Indexed: 01/02/2023]
Abstract
Bioconversion using microorganisms and their enzymes is an important tool in many industrial fields. The discovery of useful new microbial enzymes contributes to the development of industries utilizing bioprocesses. Streptomyces sp. EAS-AB2608, isolated from a soil sample collected in Japan, can convert the tetrahydrobenzotriazole CPD-1 (a selective positive allosteric modulator of metabotropic glutamate receptor 5) to its hydroxylated form at the C4-(R) position. The current study was performed to identify the genes encoding the enzymes involved in CPD-1 bioconversion and to verify their function. To identify gene products responsible for the conversion of CPD-1, we used RNA sequencing to analyze EAS-AB2608; from its 8333 coding sequences, we selected two genes, one encoding cytochrome P450 (easab2608_00800) and the other encoding ferredoxin (easab2608_00799), as encoding desirable gene products involved in the bioconversion of CPD-1. The validity of this selection was tested by using a heterologous expression approach. A bioconversion assay using genetically engineered Streptomyces avermitilis SUKA24 ∆saverm3882 ∆saverm7246 co-expressing the two selected genes (strain ES_SUKA_63) confirmed that these gene products had hydroxylation activity with respect to CPD-1, indicating that they are responsible for the conversion of CPD-1. Strain ES_SUKA_63 also showed oxidative activity toward other compounds and therefore might be useful not only for bioconversion of CPD-1 but also as a tool for synthesis of drug metabolites and in optimization studies of various pharmaceutical lead compounds. We expect that this approach will be useful for bridging the gap between the latest enzyme optimization technologies and conventional enzyme screening using microorganisms. KEY POINTS: • Genes easab2608_00800 (cyp) and easab2608_00799 (fdx) were selected by RNA-Seq. • Selection validity was evaluated by an engineered S. avermitilis expression system. • Strain ES_SUKA_63 showed oxidative activity toward CPD-1 and other compounds.
Collapse
|
27
|
Filon MJ, Wallace E, Wright S, Douglas DJ, Steinberg LI, Verkuilen CL, Westmark PR, Maganti RK, Westmark CJ. Sleep and diurnal rest-activity rhythm disturbances in a mouse model of Alzheimer's disease. Sleep 2021; 43:5830779. [PMID: 32369586 DOI: 10.1093/sleep/zsaa087] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/07/2020] [Indexed: 01/08/2023] Open
Abstract
STUDY OBJECTIVES Accumulating evidence suggests a strong association between sleep, amyloid-beta (Aβ) deposition, and Alzheimer's disease (AD). We sought to determine if (1) deficits in rest-activity rhythms and sleep are significant phenotypes in J20 AD mice, (2) metabotropic glutamate receptor 5 inhibitors (mGluR5) could rescue deficits in rest-activity rhythms and sleep, and (3) Aβ levels are responsive to treatment with mGluR5 inhibitors. METHODS Diurnal rest-activity levels were measured by actigraphy and sleep-wake patterns by electroencephalography, while animals were chronically treated with mGluR5 inhibitors. Behavioral tests were performed, and Aβ levels measured in brain lysates. RESULTS J20 mice exhibited a 4.5-h delay in the acrophase of activity levels compared to wild-type littermates and spent less time in rapid eye movement (REM) sleep during the second half of the light period. J20 mice also exhibited decreased non-rapid eye movement (NREM) delta power but increased NREM sigma power. The mGluR5 inhibitor CTEP rescued the REM sleep deficit and improved NREM delta and sigma power but did not correct rest-activity rhythms. No statistically significant differences were observed in Aβ levels, rotarod performance, or the passive avoidance task following chronic mGluR5 inhibitor treatment. CONCLUSIONS J20 mice have disruptions in rest-activity rhythms and reduced homeostatic sleep pressure (reduced NREM delta power). NREM delta power was increased following treatment with a mGluR5 inhibitor. Drug bioavailability was poor. Further work is necessary to determine if mGluR5 is a viable target for treating sleep phenotypes in AD.
Collapse
Affiliation(s)
- Mikolaj J Filon
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | - Eli Wallace
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | - Samantha Wright
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | - Dylan J Douglas
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | | | | | - Pamela R Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | - Rama K Maganti
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| | - Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
28
|
Yang W, Zhou X, Ryazanov AG, Ma T. Suppression of the kinase for elongation factor 2 alleviates mGluR-LTD impairments in a mouse model of Alzheimer's disease. Neurobiol Aging 2021; 98:225-230. [PMID: 33341653 PMCID: PMC8201868 DOI: 10.1016/j.neurobiolaging.2020.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/05/2020] [Accepted: 11/22/2020] [Indexed: 01/06/2023]
Abstract
Impaired mRNA translation (protein synthesis) is linked to Alzheimer's disease (AD) pathophysiology. Recent studies revealed the role of increased phosphorylation of eukaryotic elongation factor 2 (eEF2) in AD-associated cognitive deficits. Phosphorylation of eEF2 (at the Thr56 site) by its only known kinase eEF2K leads to inhibition of general protein synthesis. AD is considered as a disease of "synaptic failure" characterized by impairments of synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD). Deficiency of metabotropic glutamate receptor 5-dependent LTD (mGluR-LTD) is indicated in cognitive syndromes associated with various neurological disorders, including AD, but the molecular signaling mechanisms underlying the mGluR-LTD dysregulation in AD remain unclear. In this brief communication, we report genetic repression of eEF2K in aged APP/PS1 AD model mice prevented AD-associated hippocampal mGluR-LTD deficits. Using a pharmacological approach, we further observed that impairments of mGluR-LTD in APP/PS1 mice were rescued by treating hippocampal slices with a small molecule eEF2K antagonist NH125. Our findings, taken together, suggest a critical role of abnormal protein synthesis dysregulation at the elongation phase in AD-associated mGluR-LTD failure, thus providing insights into a mechanistic understanding of synaptic impairments in AD and other related dementia syndromes.
Collapse
Affiliation(s)
- Wenzhong Yang
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Xueyan Zhou
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Alexey G Ryazanov
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
29
|
Integrative analysis of genome-wide association studies identifies novel loci associated with neuropsychiatric disorders. Transl Psychiatry 2021; 11:69. [PMID: 33479212 PMCID: PMC7820351 DOI: 10.1038/s41398-020-01195-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 01/30/2023] Open
Abstract
Neuropsychiatric disorders, such as autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), schizophrenia (SCZ), bipolar disorder (BIP), and major depressive disorder (MDD) share common clinical presentations, suggesting etiologic overlap. A substantial proportion of SNP-based heritability for neuropsychiatric disorders is attributable to genetic components, and genome-wide association studies (GWASs) focusing on individual diseases have identified multiple genetic loci shared between these diseases. Here, we aimed at identifying novel genetic loci associated with individual neuropsychiatric diseases and genetic loci shared by neuropsychiatric diseases. We performed multi-trait joint analyses and meta-analysis across five neuropsychiatric disorders based on their summary statistics from the Psychiatric Genomics Consortium (PGC), and further carried out a replication study of ADHD among 2726 cases and 16299 controls in an independent pediatric cohort. In the multi-trait joint analyses, we found five novel genome-wide significant loci for ADHD, one novel locus for BIP, and ten novel loci for MDD. We further achieved modest replication in our independent pediatric dataset. We conducted fine-mapping and functional annotation through an integrative multi-omics approach and identified causal variants and potential target genes at each novel locus. Gene expression profile and gene-set enrichment analysis further suggested early developmental stage expression pattern and postsynaptic membrane compartment enrichment of candidate genes at the genome-wide significant loci of these neuropsychiatric disorders. Therefore, through a multi-omics approach, we identified novel genetic loci associated with the five neuropsychiatric disorders which may help to better understand the underlying molecular mechanism of neuropsychiatric diseases.
Collapse
|
30
|
Effects of blocking mGluR5 on primate dorsolateral prefrontal cortical neuronal firing and working memory performance. Psychopharmacology (Berl) 2021; 238:97-106. [PMID: 32939596 PMCID: PMC7794104 DOI: 10.1007/s00213-020-05661-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022]
Abstract
RATIONALE Metabotropic glutamate type 5 receptor (mGluR5) antagonists are under development for treating cognitive disorders such as Fragile X syndrome and Alzheimer's disease, largely based on success in mouse models, where post-synaptic mGluR5 stimulation weakens synaptic functions in hippocampus. However, human trials of mGluR5 antagonists have yet to be successful. This may be due in part to the differing effects of mGluR5 in hippocampus vs. prefrontal cortex, as mGluR5 are primarily post-synaptic in rodent hippocampus, but are both pre- and post-synaptic in the dorsolateral prefrontal cortical (dlPFC) circuits known to subserve working memory. OBJECTIVES AND METHODS The current study examined the effects of the selective mGluR5 negative allosteric modulator, MTEP (3-((2-Methyl-1,3-thiazol-4-yl)ethynyl)pyridine hydrochloride), on neuronal firing and working memory performance in aging rhesus monkeys with naturally occurring impairments in neuronal firing and cognitive performance. RESULTS We found that iontophoresis of MTEP directly onto dlPFC "Delay cells" had an inverted U dose-response, where low doses tended to enhance task-related firing, but higher doses suppressed neuronal firing. Similar effects were seen on cognitive performance following systemic MTEP administration (0.0001-0.1 mg/kg), with MTEP producing erratic dose-response curves. In the subset of monkeys (50%) that showed replicable improvement with MTEP, co-administration with the mGluR5 PAM, CDPPB (3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide), blocked MTEP beneficial effects, consistent with mGluR5 actions. CONCLUSIONS The mixed effects of MTEP on cognitive performance may arise from opposing actions at pre- vs. post-synaptic mGluR5 in dlPFC. These data from monkeys suggest that future clinical trials should include low doses, and identification of potential subgroup responders.
Collapse
|
31
|
Kim JH, Marton J, Ametamey SM, Cumming P. A Review of Molecular Imaging of Glutamate Receptors. Molecules 2020; 25:molecules25204749. [PMID: 33081223 PMCID: PMC7587586 DOI: 10.3390/molecules25204749] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Molecular imaging with positron emission tomography (PET) and single photon emission computed tomography (SPECT) is a well-established and important in vivo technique to evaluate fundamental biological processes and unravel the role of neurotransmitter receptors in various neuropsychiatric disorders. Specific ligands are available for PET/SPECT studies of dopamine, serotonin, and opiate receptors, but corresponding development of radiotracers for receptors of glutamate, the main excitatory neurotransmitter in mammalian brain, has lagged behind. This state of affairs has persisted despite the central importance of glutamate neurotransmission in brain physiology and in disorders such as stroke, epilepsy, schizophrenia, and neurodegenerative diseases. Recent years have seen extensive efforts to develop useful ligands for molecular imaging of subtypes of the ionotropic (N-methyl-D-aspartate (NMDA), kainate, and AMPA/quisqualate receptors) and metabotropic glutamate receptors (types I, II, and III mGluRs). We now review the state of development of radioligands for glutamate receptor imaging, placing main emphasis on the suitability of available ligands for reliable in vivo applications. We give a brief account of the radiosynthetic approach for selected molecules. In general, with the exception of ligands for the GluN2B subunit of NMDA receptors, there has been little success in developing radiotracers for imaging ionotropic glutamate receptors; failure of ligands for the PCP/MK801 binding site in vivo doubtless relates their dependence on the open, unblocked state of the ion channel. Many AMPA and kainite receptor ligands with good binding properties in vitro have failed to give measurable specific binding in the living brain. This may reflect the challenge of developing brain-penetrating ligands for amino acid receptors, compounded by conformational differences in vivo. The situation is better with respect to mGluR imaging, particularly for the mGluR5 subtype. Several successful PET ligands serve for investigations of mGluRs in conditions such as schizophrenia, depression, substance abuse and aging. Considering the centrality and diversity of glutamatergic signaling in brain function, we have relatively few selective and sensitive tools for molecular imaging of ionotropic and metabotropic glutamate receptors. Further radiopharmaceutical research targeting specific subtypes and subunits of the glutamate receptors may yet open up new investigational vistas with broad applications in basic and clinical research.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Incheon 21565, Korea
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| | - János Marton
- ABX Advanced Biochemical Compounds, Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-1454 Radeberg, Germany;
| | - Simon Mensah Ametamey
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland;
| | - Paul Cumming
- Department of Nuclear Medicine, University of Bern, Inselspital, Freiburgstrasse 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane QLD 4059, Australia
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| |
Collapse
|
32
|
Sánchez-Melgar A, Albasanz JL, Pallàs M, Martín M. Resveratrol Differently Modulates Group I Metabotropic Glutamate Receptors Depending on Age in SAMP8 Mice. ACS Chem Neurosci 2020; 11:1770-1780. [PMID: 32437602 DOI: 10.1021/acschemneuro.0c00067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glutamate homeostasis is critical for neurotransmission as this excitatory neurotransmitter has a relevant role in cognition functions through ionotropic and metabotropic glutamate receptors in the central nervous system. During the last years, the role of the group I metabotropic glutamate receptors (mGluRs) in neurodegenerative diseases such as Alzheimer's disease has been intensely investigated. Resveratrol (RSV) is a natural polyphenolic compound that is thought to have neuroprotective properties for human health. However, little is known about the action of this compound on mGluR signaling. Therefore, the aim of this study was to investigate the possible modulation of group I mGluRs in SAMP8 mice five and seven months of age supplemented with RSV in the diet. Data reported herein show that RSV plays a different modulatory action on group I mGluRs: mGluR5 is downregulated as age increases, independently of RSV presence, and mGluR1 is upregulated or downregulated by RSV treatment depending on age (i.e., depending on mGluR5 levels). In addition, a neuroprotective role can be inferred for RSV as lower glutamate levels, higher synapsin levels, and unchanged caspase-3 activity were detected after RSV treatment. In conclusion, our findings indicate that RSV treatment modifies the group I mGluR-mediated glutamatergic system in SAMP8 mice, which could contribute to the beneficial effects of this natural polyphenol.
Collapse
Affiliation(s)
- Alejandro Sánchez-Melgar
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical Sciences and Technologies, Faculty of Medicine of Ciudad Real, Regional Center of Biomedical Research, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - José Luis Albasanz
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical Sciences and Technologies, Faculty of Medicine of Ciudad Real, Regional Center of Biomedical Research, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Mercé Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona, Barcelona 08024, Spain
| | - Mairena Martín
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Chemical Sciences and Technologies, Faculty of Medicine of Ciudad Real, Regional Center of Biomedical Research, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| |
Collapse
|
33
|
Zhang X, Lao K, Qiu Z, Rahman MS, Zhang Y, Gou X. Potential Astrocytic Receptors and Transporters in the Pathogenesis of Alzheimer's Disease. J Alzheimers Dis 2020; 67:1109-1122. [PMID: 30741675 DOI: 10.3233/jad-181084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is characterized by the progressive loss of memory and cognition in the aging population. However, the etiology of and therapies for AD remain far from understood. Astrocytes, the most abundant neuroglia in the brain, have recently aroused substantial concern due to their involvement in synaptotoxicity, amyloidosis, neuroinflammation, and oxidative stress. In this review, we summarize the candidate molecules of astrocytes, especially receptors and transporters, that may be involved in AD pathogenesis. These molecules include excitatory amino acid transporters (EAATs), metabotropic glutamate receptor 5 (mGluR5), the adenosine 2A receptor (A2AR), the α7-nicotinic acetylcholine receptor (α7-nAChR), the calcium-sensing receptor (CaSR), S100β, and cannabinoid receptors. We describe the characteristics of these molecules and the neurological and pharmacological underpinnings of these molecules in AD. Among these molecules, EAATs, A2AR, and mGluR5 are strongly related to glutamate-mediated synaptotoxicity and are involved in glutamate transmission or the clearance of extrasynaptic glutamate in the AD brain. The α7-nAChR, CaSR, and mGluR5 are receptors of Aβ and can induce a plethora of toxic effects, such as the production of excess Aβ, synaptotoxicity, and NO production triggered by changes in intracellular calcium signaling. Antagonists or positive allosteric modulators of these receptors can repair cognitive ability and modify neurobiological changes. Moreover, blocking S100β or activating cannabinoid receptors reduces neuroinflammation, oxidative stress, and reactive astrogliosis. Thus, targeting these molecules might provide alternative approaches for treating AD.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China
| | - Kejing Lao
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China
| | - Zhongying Qiu
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China
| | - Md Saidur Rahman
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China.,Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Yuelin Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, P.R. China
| |
Collapse
|
34
|
JBPOS0101 attenuates amyloid-β accumulation and memory loss in a mouse model of Alzheimer's disease. Neuroreport 2020; 30:741-747. [PMID: 31095107 DOI: 10.1097/wnr.0000000000001269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) in the brain. Defects in Aβ clearance or the interference of Aβ homeostasis could result in Aβ aggregation. JBPOS0101 has been studied for its antiepileptic activity. It showed a neuroprotective effect and prevented memory deficits in lithium-pilocarpine-induced status epilepticus rats. In this study, we tested the effect of JBPOS0101 in an AD model. We showed that JBPOS0101 attenuated the accumulation of Aβ in 5XFAD mouse brains. Moreover, the treatment of JBPOS0101 rescued the deficits in learning and memory in 5XFAD mice. These data suggest that JBPOS0101 could be a potential therapeutic drug candidate for AD.
Collapse
|
35
|
Zhang X, Wang D, Zhang B, Zhu J, Zhou Z, Cui L. Regulation of microglia by glutamate and its signal pathway in neurodegenerative diseases. Drug Discov Today 2020; 25:1074-1085. [PMID: 32320851 DOI: 10.1016/j.drudis.2020.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/10/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Microglia are an essential component of the central nervous system (CNS) and are involved in the primary response to microorganisms, neuroinflammation, homeostasis, and tissue regeneration, as well as contributing to the pathogenesis of neurodegenerative diseases. Research has shown that microglial diversity, multifunctionality, and their relationship with glutamate are crucial to determining their roles in these diseases. In this review, we focus on recent progress in determining microglial characteristics and the role of glutamate and its receptors in microglia regulation, which could be a novel therapeutic strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Dan Wang
- Department of Ophthalmology, the First Hospital of Jilin University, Changchun, China.
| | - Bo Zhang
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden; Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China.
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Zhulin Zhou
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
36
|
Chronic Activation of Gp1 mGluRs Leads to Distinct Refinement of Neural Network Activity through Non-Canonical p53 and Akt Signaling. eNeuro 2020; 7:ENEURO.0438-19.2020. [PMID: 32161037 PMCID: PMC7218008 DOI: 10.1523/eneuro.0438-19.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/17/2020] [Accepted: 03/02/2020] [Indexed: 11/30/2022] Open
Abstract
Group 1 metabotropic glutamate receptors (Gp1 mGluRs), including mGluR1 and mGluR5, are critical regulators for neuronal and synaptic plasticity. Dysregulated Gp1 mGluR signaling is observed with various neurologic disorders, including Alzheimer’s disease, Parkinson’s disease, epilepsy, and autism spectrum disorders (ASDs). It is well established that acute activation of Gp1 mGluRs leads to elevation of neuronal intrinsic excitability and long-term synaptic depression. However, it remains unknown how chronic activation of Gp1 mGluRs can affect neural activity and what molecular mechanisms might be involved. In the current study, we employed a multielectrode array (MEA) recording system to evaluate neural network activity of primary mouse cortical neuron cultures. We demonstrated that chronic activation of Gp1 mGluRs leads to elevation of spontaneous spike frequency while burst activity and cross-electrode synchronization are maintained at the baseline. We further showed that these neural network properties are achieved through proteasomal degradation of Akt that is dependent on the tumor suppressor p53. Genetically knocking down p53 disrupts the elevation of spontaneous spike frequency and alters the burst activity and cross-electrode synchronization following chronic activation of Gp1 mGluRs. Importantly, these deficits can be restored by pharmacologically inhibiting Akt to mimic inactivation of Akt mediated by p53. Together, our findings reveal the effects of chronic activation of Gp1 mGluRs on neural network activity and identify a unique signaling pathway involving p53 and Akt for these effects. Our data can provide insights into constitutively active Gp1 mGluR signaling observed in many neurologic and psychiatric disorders.
Collapse
|
37
|
Varlow C, Murrell E, Holland JP, Kassenbrock A, Shannon W, Liang SH, Vasdev N, Stephenson NA. Revisiting the Radiosynthesis of [ 18F]FPEB and Preliminary PET Imaging in a Mouse Model of Alzheimer's Disease. Molecules 2020; 25:molecules25040982. [PMID: 32098347 PMCID: PMC7070414 DOI: 10.3390/molecules25040982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/24/2022] Open
Abstract
[18F]FPEB is a positron emission tomography (PET) radiopharmaceutical used for imaging the abundance and distribution of mGluR5 in the central nervous system (CNS). Efficient radiolabeling of the aromatic ring of [18F]FPEB has been an ongoing challenge. Herein, five metal-free precursors for the radiofluorination of [18F]FPEB were compared, namely, a chloro-, nitro-, sulfonium salt, and two spirocyclic iodonium ylide (SCIDY) precursors bearing a cyclopentyl (SPI5) and a new adamantyl (SPIAd) auxiliary. The chloro- and nitro-precursors resulted in a low radiochemical yield (<10% RCY), whereas both SCIDY precursors and the sulfonium salt precursor produced [18F]FPEB in the highest RCYs of 25% and 36%, respectively. Preliminary PET/CT imaging studies with [18F]FPEB were conducted in a transgenic model of Alzheimer’s Disease (AD) using B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J (APP/PS1) mice, and data were compared with age-matched wild-type (WT) B6C3F1/J control mice. In APP/PS1 mice, whole brain distribution at 5 min post-injection showed a slightly higher uptake (SUV = 4.8 ± 0.4) than in age-matched controls (SUV = 4.0 ± 0.2). Further studies to explore mGluR5 as an early biomarker for AD are underway.
Collapse
Affiliation(s)
- Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
| | - Jason P. Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Chemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Alina Kassenbrock
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
| | - Whitney Shannon
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N OX2, Canada
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T-1R8, Canada
- Correspondence: (N.V.); (N.A.S.); Tel.: +416-535-8501 (ext. 30988) (N.V.); +1-876-927-1910 (N.A.S.)
| | - Nickeisha A. Stephenson
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (C.V.); (E.M.); (W.S.)
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; (J.P.H.); (A.K.); (S.H.L.)
- Department of Chemistry, The University of West Indies at Mona, Kingston 7, Jamaica
- Correspondence: (N.V.); (N.A.S.); Tel.: +416-535-8501 (ext. 30988) (N.V.); +1-876-927-1910 (N.A.S.)
| |
Collapse
|
38
|
Zhang J, Zhang F, Wu J, Li J, Yang Z, Yue J. Glutamate affects cholesterol homeostasis within the brain via the up-regulation of CYP46A1 and ApoE. Toxicology 2020; 432:152381. [PMID: 31981724 DOI: 10.1016/j.tox.2020.152381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
Abstract
Chronic glutamate excitotoxicity has been thought to be involved in numerous neurodegenerative disorders. A small but significant loss of membrane cholesterol has been reported following a short stimulation of ionotropic glutamate receptors (iGluRs). We investigated the alteration of brain cholesterol following chronic glutamate treatment. The alteration of cholesterol levels was evaluated in the hippocampus from the adult rats that received the subcutaneous injection with monosodium l-glutamate at 1, 3, 5, and 7 days of age. The regulation of CYP46A1, LXRα, and ApoE levels were assayed following subtoxic glutamate treatment in SH-SY5Y cells as well as HT-22 cells lacking iGluRs. The ratio of 24S-hydroxycholesterol to cholesterol was elevated in the adult rats exposed to monosodium l-glutamate before the weaning age, compared to the control. The blockers of NMDA receptor (MK801) and mGluR5 (MPEP) attenuated the glutamate-induced loss of cholesterol and elevation of 24S-hydroxycholesterol level in SH-SY5Y cells. The induction of the mRNA levels of CYP46A1, LXRα, and ApoE by glutamate was observed in both SH-SY5Y cells and HT-22 cells; additionally, MK801 and MPEP attenuated the increases in these genes in SH-SY5Y cells. The increase in the binding of LXRα proteins with ApoE promoter following glutamate treatment was attenuated by MK801. The luciferase assay indicated the binding of CREB protein with CYP46A1 promoter, and the glutamate-induced CREB expression was inhibited by MK801. The results suggest that glutamate, the major excitatory neurotransmitter, may affect the metabolism and redistribution of cholesterol in the neuronal cells via its specific receptors during chronic exposure.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Department of Clinical Pharmacology, PLA General Hospital of Central Theater Command, Wuhan 430061, China
| | - Furong Zhang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Juan Wu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jie Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Zheqiong Yang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jiang Yue
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Medical Research Center for Structural Biology, Basic Medical School of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
39
|
Srivastava A, Das B, Yao AY, Yan R. Metabotropic Glutamate Receptors in Alzheimer's Disease Synaptic Dysfunction: Therapeutic Opportunities and Hope for the Future. J Alzheimers Dis 2020; 78:1345-1361. [PMID: 33325389 PMCID: PMC8439550 DOI: 10.3233/jad-201146] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the presence of neuritic plaques and neurofibrillary tangles. The impaired synaptic plasticity and dendritic loss at the synaptic level is an early event associated with the AD pathogenesis. The abnormal accumulation of soluble oligomeric amyloid-β (Aβ), the major toxic component in amyloid plaques, is viewed to trigger synaptic dysfunctions through binding to several presynaptic and postsynaptic partners and thus to disrupt synaptic transmission. Over time, the abnormalities in neural transmission will result in cognitive deficits, which are commonly manifested as memory loss in AD patients. Synaptic plasticity is regulated through glutamate transmission, which is mediated by various glutamate receptors. Here we review recent progresses in the study of metabotropic glutamate receptors (mGluRs) in AD cognition. We will discuss the role of mGluRs in synaptic plasticity and their modulation as a possible strategy for AD cognitive improvement.
Collapse
Affiliation(s)
- Akriti Srivastava
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Annie Y. Yao
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
40
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [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: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
41
|
Sushma, Mondal AC. Role of GPCR signaling and calcium dysregulation in Alzheimer's disease. Mol Cell Neurosci 2019; 101:103414. [PMID: 31655116 DOI: 10.1016/j.mcn.2019.103414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), a late onset neurodegenerative disorder is characterized by the loss of memory, disordered cognitive function, caused by accumulation of amyloid-β (Aβ) peptide and neurofibrillary tangles (NFTs) in the neocortex and hippocampal brain area. Extensive research has been done on the findings of the disease etiology or pathological causes of aggregation of Aβ and hyperphosphorylation of tau protein without much promising results. Recently, calcium dysregulation has been reported to play an important role in the pathophysiology of AD. Calcium ion acts as one of the major secondary messengers, regulates many signaling pathways involved in cell survival, proliferation, differentiation, transcription and apoptosis. Calcium signaling is one of the major signaling pathways involved in the formation of memory, generation of energy and other physiological functions. It also can modulate function of many proteins upon binding. Dysregulation in calcium homeostasis leads to many physiological changes leading to neurodegenerative diseases including AD. In AD, GPCRs generate secondary messengers which regulate calcium homeostasis inside the cell and is reported to be disturbed in the pathological condition. Calcium channels and receptors present on the plasma membrane and intracellular organelle maintain calcium homeostasis through different signaling mechanisms. In this review, we have summarized the different calcium channels and receptors involved in calcium dysregulation which in turn play a critical role in the pathogenesis of AD. Understanding the role of calcium channels and GPCRs to maintain calcium homeostasis is an attempt to develop effective AD treatments.
Collapse
Affiliation(s)
- Sushma
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
42
|
Bellozi PMQ, Gomes GF, da Silva MCM, Lima IVDA, Batista CRÁ, Carneiro Junior WDO, Dória JG, Vieira ÉLM, Vieira RP, de Freitas RP, Ferreira CN, Candelario-Jalil E, Wyss-Coray T, Ribeiro FM, de Oliveira ACP. A positive allosteric modulator of mGluR5 promotes neuroprotective effects in mouse models of Alzheimer's disease. Neuropharmacology 2019; 160:107785. [PMID: 31541651 DOI: 10.1016/j.neuropharm.2019.107785] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/06/2019] [Accepted: 09/17/2019] [Indexed: 12/26/2022]
Abstract
Alzheimer's Disease (AD) is the most prevalent neurodegenerative disorder. Despite advances in the understanding of its pathophysiology, none of the available therapies prevents disease progression. Excess glutamate plays an important role in excitotoxicity by activating ionotropic receptors. However, the mechanisms modulating neuronal cell survival/death via metabotropic glutamate receptors (mGluRs) are not completely understood. Recent data indicates that CDPPB, a positive allosteric modulator of mGluR5, has neuroprotective effects. Thus, this work aimed to investigate CDPPB treatment effects on amyloid-β (Aβ) induced pathological alterations in vitro and in vivo and in a transgenic mouse model of AD (T41 mice). Aβ induced cell death in primary cultures of hippocampal neurons, which was prevented by CDPPB. Male C57BL/6 mice underwent stereotaxic surgery for unilateral intra-hippocampal Aβ injection, which induced memory deficits, neurodegeneration, neuronal viability reduction and decrease of doublecortin-positive cells, a marker of immature neurons and neuronal proliferation. Treatment with CDPPB for 8 days reversed neurodegeneration and doublecortin-positive cells loss and recovered memory function. Fourteen months old T41 mice presented cognitive deficits, neuronal viability reduction, gliosis and Aβ accumulation. Treatment with CDPPB for 28 days increased neuronal viability (32.2% increase in NeuN+ cells) and reduced gliosis in CA1 region (Iba-1+ area by 31.3% and GFAP+ area by 37.5%) in transgenic animals, without inducing hepatotoxicity. However, it did not reverse cognitive deficit. Despite a four-week treatment did not prevent memory loss in aged transgenic mice, CDPPB is protective against Aβ stimulus. Therefore, this drug represents a potential candidate for further investigations as AD treatment.
Collapse
Affiliation(s)
| | - Giovanni Freitas Gomes
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | | | | | | | - Juliana Guimarães Dória
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Rafael Pinto Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Claudia Natália Ferreira
- Clinical Pathology Sector of COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | |
Collapse
|
43
|
Forest KH, Nichols RA. Assessing Neuroprotective Agents for Aβ-Induced Neurotoxicity. Trends Mol Med 2019; 25:685-695. [DOI: 10.1016/j.molmed.2019.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/17/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
|
44
|
Nagayach A, Singh A, Geller AI. Separate Gene Transfers into Pre- and Postsynaptic Neocortical Neurons Connected by mGluR5-Containing Synapses. J Mol Neurosci 2019; 68:549-564. [PMID: 30972540 PMCID: PMC6615967 DOI: 10.1007/s12031-019-01317-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/28/2019] [Indexed: 11/25/2022]
Abstract
mGluR5-containing synapses have essential roles in synaptic plasticity, circuit physiology, and learning, and dysfunction at these synapses is implicated in specific neurological disorders. As mGluR5-containing synapses are embedded in large and complex distributed circuits containing many neuron and synapse types, it is challenging to elucidate the roles of these synapses and to develop treatments for the associated disorders. Thus, it would be advantageous to deliver different genes into pre- and postsynaptic neurons connected by a mGluR5-containing synapse. Here, we develop this capability: The first gene transfer, into the presynaptic neurons, uses standard techniques to deliver a vector that expresses a synthetic peptide neurotransmitter. This peptide neurotransmitter has three domains: a dense core vesicle sorting domain, a mGluR5-binding domain composed of a single-chain variable fragment anti-mGluR5, and the His tag. Upon release, this peptide neurotransmitter binds to mGluR5, predominately located on the postsynaptic neurons. Selective gene transfer into these neurons uses antibody-mediated, targeted gene transfer and anti-His tag antibodies, as the synthetic peptide neurotransmitter contains the His tag. For the model system, we studied the connection between neurons in two neocortical areas: postrhinal and perirhinal cortices. Targeted gene transfer was over 80% specific for mGluR5-containing synapses, but untargeted gene transfer was only ~ 15% specific for these synapses. This technology may enable studies on the roles of mGluR5-containing neurons and synapses in circuit physiology and learning and support gene therapy treatments for specific disorders that involve dysfunction at these synapses.
Collapse
Affiliation(s)
- Aarti Nagayach
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Anshuman Singh
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Alfred I Geller
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, USA.
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, USA.
| |
Collapse
|
45
|
Alzheimer's disease: Key developments support promising perspectives for therapy. Pharmacol Res 2019; 146:104316. [PMID: 31260730 DOI: 10.1016/j.phrs.2019.104316] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/15/2019] [Accepted: 06/15/2019] [Indexed: 12/13/2022]
Abstract
Alzheimer's is the neurodegenerative disease affecting the largest number of patients in the world. In spite of the intense research of the last decades, progress about its knowledge and therapy was limited. In particular, various cytotoxic processes remained debated, while the few drugs approved for therapy were of only marginal relevance. Recent studies have identified key aspects of the disease, such as the mechanisms governing the development of pathology. In order to operate the Aβ peptide, known as the key factor, requires a complex assembled by its high affinity binding to PrPc, a cell surface prion protein, and mGluR5, a metabotropic glutamate receptor. Aβ and its associates bind also phosphorylated tau transferred to the extracellular space, with final activation of intracellular cytotoxic signals. Pathology is further affected by factors (including genes, receptors and their agonists) and by glial cells governing (via vesicles, cytokines and enzymes) cell immunology, inflammation and oxidative stress. Concomitant to pathology studies, strong attempts have been made for the development of new, effective therapies. Critical for this are biomarkers, by which Alzheimer's patients are recognized even before appearance of their symptoms. The question was whether patients take advantage from drugs not yet approved. The latter, first identified in mice, were found effective also in men, however only before appearance or at early stage of the disease. In other words, the drugs not yet approved induce effective protection of patients still healthy or in a preliminary stage of the disease. In contrast, developed Alzheimer's disease is practically irreversible.
Collapse
|
46
|
Chen Y, Fu AKY, Ip NY. Synaptic dysfunction in Alzheimer's disease: Mechanisms and therapeutic strategies. Pharmacol Ther 2018; 195:186-198. [PMID: 30439458 DOI: 10.1016/j.pharmthera.2018.11.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disease in the elderly population, is characterized by progressive cognitive decline and pathological hallmarks of amyloid plaques and neurofibrillary tangles. However, its pathophysiological mechanisms are poorly understood, and diagnostic tools and interventions are limited. Here, we review recent research on the amyloid hypothesis and beta-amyloid-induced dysfunction of neuronal synapses through distinct cell surface receptors. We also review how tau protein leads to synaptotoxicity through pathological modification, localization, and propagation. Finally, we discuss experimental therapeutics for AD and propose potential applications of disease-modifying strategies targeting synaptic failure for improved treatment of AD.
Collapse
Affiliation(s)
- Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
| |
Collapse
|
47
|
Metabotropic glutamate receptor subtype 5 is altered in LPS-induced murine neuroinflammation model and in the brains of AD and ALS patients. Eur J Nucl Med Mol Imaging 2018; 46:407-420. [PMID: 30291374 DOI: 10.1007/s00259-018-4179-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/20/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE The aim of the present study was to determine the expression levels of mGluR5 in different mouse strains after induction of neuroinflammation by lipopolysaccharide (LPS) challenge and in the brains of patients with Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS) post mortem to investigate mGluR5 expression in human neurodegenerative diseases. METHODS C57BL/6 and CD1 mice were injected intraperitoneally with either 10 mg/kg LPS or saline. mGluR5 and TSPO mRNA levels were measured after 1 and 5 days by qPCR, and mGluR5 protein levels were determined by PET imaging with the mGluR5-specific radiotracer [18F]PSS232. mGluR5 expression was evaluated in the post-mortem brain slices from AD and ALS patients using in vitro autoradiography. RESULTS mGluR5 and TSPO mRNA levels were increased in brains of C57BL/6 and CD1 mice 1 day after LPS treatment and remained significantly increased after 5 days in C57BL/6 mice but not in CD1 mice. Brain PET imaging with [18F]PSS232 confirmed increased mGluR5 levels in the brains of both mouse strains 1 day after LPS treatment. After 5 days, mGluR5 levels in CD1 mice declined to the levels in vehicle-treated mice but remained high in C57BL/6 mice. Autoradiograms revealed a severalfold higher binding of [18F]PSS232 in post-mortem brain slices from AD and ALS patients compared with the binding in control brains. CONCLUSION LPS-induced neuroinflammation increased mGluR5 levels in mouse brain and is dependent on the mouse strain and time after LPS treatment. mGluR5 levels were also increased in human AD and ALS brains in vitro. PET imaging of mGluR5 levels could potentially be used to diagnose and monitor therapy outcomes in patients with AD and ALS.
Collapse
|
48
|
Kerr F, Bjedov I, Sofola-Adesakin O. Molecular Mechanisms of Lithium Action: Switching the Light on Multiple Targets for Dementia Using Animal Models. Front Mol Neurosci 2018; 11:297. [PMID: 30210290 PMCID: PMC6121012 DOI: 10.3389/fnmol.2018.00297] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Lithium has long been used for the treatment of psychiatric disorders, due to its robust beneficial effect as a mood stabilizing drug. Lithium’s effectiveness for improving neurological function is therefore well-described, stimulating the investigation of its potential use in several neurodegenerative conditions including Alzheimer’s (AD), Parkinson’s (PD) and Huntington’s (HD) diseases. A narrow therapeutic window for these effects, however, has led to concerted efforts to understand the molecular mechanisms of lithium action in the brain, in order to develop more selective treatments that harness its neuroprotective potential whilst limiting contraindications. Animal models have proven pivotal in these studies, with lithium displaying advantageous effects on behavior across species, including worms (C. elegans), zebrafish (Danio rerio), fruit flies (Drosophila melanogaster) and rodents. Due to their susceptibility to genetic manipulation, functional genomic analyses in these model organisms have provided evidence for the main molecular determinants of lithium action, including inhibition of inositol monophosphatase (IMPA) and glycogen synthase kinase-3 (GSK-3). Accumulating pre-clinical evidence has indeed provided a basis for research into the therapeutic use of lithium for the treatment of dementia, an area of medical priority due to its increasing global impact and lack of disease-modifying drugs. Although lithium has been extensively described to prevent AD-associated amyloid and tau pathologies, this review article will focus on generic mechanisms by which lithium preserves neuronal function and improves memory in animal models of dementia. Of these, evidence from worms, flies and mice points to GSK-3 as the most robust mediator of lithium’s neuro-protective effect, but it’s interaction with downstream pathways, including Wnt/β-catenin, CREB/brain-derived neurotrophic factor (BDNF), nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and toll-like receptor 4 (TLR4)/nuclear factor-κB (NFκB), have identified multiple targets for development of drugs which harness lithium’s neurogenic, cytoprotective, synaptic maintenance, anti-oxidant, anti-inflammatory and protein homeostasis properties, in addition to more potent and selective GSK-3 inhibitors. Lithium, therefore, has advantages as a multi-functional therapy to combat the complex molecular pathology of dementia. Animal studies will be vital, however, for comparative analyses to determine which of these defense mechanisms are most required to slow-down cognitive decline in dementia, and whether combination therapies can synergize systems to exploit lithium’s neuro-protective power while avoiding deleterious toxicity.
Collapse
Affiliation(s)
- Fiona Kerr
- Department of Life Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Ivana Bjedov
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Oyinkan Sofola-Adesakin
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
49
|
Spatola M, Sabater L, Planagumà J, Martínez-Hernandez E, Armangué T, Prüss H, Iizuka T, Caparó Oblitas RL, Antoine JC, Li R, Heaney N, Tubridy N, Munteis Olivas E, Rosenfeld MR, Graus F, Dalmau J. Encephalitis with mGluR5 antibodies: Symptoms and antibody effects. Neurology 2018; 90:e1964-e1972. [PMID: 29703767 DOI: 10.1212/wnl.0000000000005614] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/09/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To report the clinical features of 11 patients with metabotropic glutamate receptor 5 (mGluR5) antibody-associated encephalitis, immunoglobulin G (IgG) subclass, and effects of the antibodies on neuronal mGluR5 clusters. METHODS Clinical information was retrospectively obtained from referring physicians. Antibodies to mGluR5 and IgG subclasses were determined with brain immunohistochemistry and cell-based assays. The effects of the antibodies were examined on rat hippocampal neurons with reported techniques. RESULTS From January 2005 to May 2017, 11 patients (median age 29 years, range 6-75 years, 5 female) were identified. The main clinical features were psychiatric (10), cognitive (10), movement disorders (7), sleep dysfunction (7), and seizures (6). Median modified Rankin Scale score at the peak of the disease was 4; 4 patients required intensive care. Five patients had Hodgkin lymphoma, and 1 had small cell lung cancer. CSF showed pleocytosis (median white blood cell count 22 mm3) in all patients; brain MRI was abnormal in 5, involving limbic (1) or extralimbic (4) regions. Treatments included immunotherapy and/or oncologic therapy; at the last follow-up (median 48 months), 6 patients had complete and 5 had partial recovery. Neurologic relapse occurred in 2 patients. Antibodies were IgG1 alone (4 of 9) or in combination with IgG2 (1 of 9), IgG3 (3 of 9), or both (1). Patients' IgG caused a significant and specific decrease of cell-surface synaptic and extrasynaptic mGluR5 without altering the levels of postsynaptic density protein 95. CONCLUSIONS Anti-mGluR5 encephalitis associates with a complex neuropsychiatric syndrome, not restricted to limbic encephalitis, and can occur without tumor. Patients respond to treatment, but relapses can occur. The antibodies have pathogenic effects altering the levels of cell-surface mGluR5.
Collapse
Affiliation(s)
- Marianna Spatola
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Lidia Sabater
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Jesús Planagumà
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Eugenia Martínez-Hernandez
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Thaís Armangué
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Harald Prüss
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Takahiro Iizuka
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Ruben L Caparó Oblitas
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Jean-Christophe Antoine
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Richard Li
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Nicholas Heaney
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Niall Tubridy
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Elvira Munteis Olivas
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Myrna R Rosenfeld
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Francesc Graus
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain
| | - Josep Dalmau
- From the Institut d'Investigacions Biomèdiques August Pi i Sunyer (M.S., L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.) and Hospital Clínic, University of Barcelona, Spain; Department of Clinical Neuroscience (M.S.), University of Lausanne, Switzerland; Centro de Investigación Biomédica en Red de Enfermedades Raras (L.S., J.P., E.M.-H., T.A., M.R.R., F.G., J.D.), Valencia; ICFO-Institut de Ciències Fotòniques (J.P.); Pediatric Neuroimmunology Unit (T.A.), Sant Joan de Déu Children's Hospital, University of Barcelona, Spain; Charité Universitätsmedizin Berlin (H.P.), Experimentelle Neurologie und Klinik und Poliklinik für Neurologie; German Center for Neurodegenerative Diseases (H.P.), Berlin, Germany; Department of Neurology (T.I.), Kitasato University School of Medicine, Sagamihara, Japan; Hospital Nacional Edgardo Rebagliati Martins (R.L.C.O.), Lima, Peru; Department of Neurology (J.-C.A.), University Hospital, Saint-Etienne, France; Pamela Youde Nethersole Eastern Hospital (R.L.), Hong Kong; Beatson West of Scotland Cancer Centre (N.H.), Glasgow, UK; Department of Neurology (N.T.), St. Vincent's University Hospital, Dublin, Ireland; Service of Neurology (E.M.O.), Hospital del Mar, IMIM, Barcelona, Spain; Department of Neurology (M.R.R., J.D.), University of Pennsylvania, Philadelphia; and Catalan Institution for Research and Advanced Studies (J.D.), Barcelona, Spain.
| |
Collapse
|
50
|
Liu DC, Seimetz J, Lee KY, Kalsotra A, Chung HJ, Lu H, Tsai NP. Mdm2 mediates FMRP- and Gp1 mGluR-dependent protein translation and neural network activity. Hum Mol Genet 2018; 26:3895-3908. [PMID: 29016848 DOI: 10.1093/hmg/ddx276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Activating Group 1 (Gp1) metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, elicits translation-dependent neural plasticity mechanisms that are crucial to animal behavior and circuit development. Dysregulated Gp1 mGluR signaling has been observed in numerous neurological and psychiatric disorders. However, the molecular pathways underlying Gp1 mGluR-dependent plasticity mechanisms are complex and have been elusive. In this study, we identified a novel mechanism through which Gp1 mGluR mediates protein translation and neural plasticity. Using a multi-electrode array (MEA) recording system, we showed that activating Gp1 mGluR elevates neural network activity, as demonstrated by increased spontaneous spike frequency and burst activity. Importantly, we validated that elevating neural network activity requires protein translation and is dependent on fragile X mental retardation protein (FMRP), the protein that is deficient in the most common inherited form of mental retardation and autism, fragile X syndrome (FXS). In an effort to determine the mechanism by which FMRP mediates protein translation and neural network activity, we demonstrated that a ubiquitin E3 ligase, murine double minute-2 (Mdm2), is required for Gp1 mGluR-induced translation and neural network activity. Our data showed that Mdm2 acts as a translation suppressor, and FMRP is required for its ubiquitination and down-regulation upon Gp1 mGluR activation. These data revealed a novel mechanism by which Gp1 mGluR and FMRP mediate protein translation and neural network activity, potentially through de-repressing Mdm2. Our results also introduce an alternative way for understanding altered protein translation and brain circuit excitability associated with Gp1 mGluR in neurological diseases such as FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program
| | - Joseph Seimetz
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology
| | - Auinash Kalsotra
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,Carl R.Woese Institute of Genomic Biology, University of Illinois, Champaign, IL 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|