1
|
Sherif MA, Carter WG, Mellor IR. Chlorpyrifos Acts as a Positive Modulator and an Agonist of N-Methyl-d-Aspartate (NMDA) Receptors: A Novel Mechanism of Chlorpyrifos-Induced Neurotoxicity. J Xenobiot 2025; 15:12. [PMID: 39846544 PMCID: PMC11755529 DOI: 10.3390/jox15010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Chlorpyrifos (CPF) is a broad-spectrum organophosphate insecticide. Long-term exposure to low levels of CPF is associated with neurodevelopmental and neurodegenerative disorders. The mechanisms leading to these effects are still not fully understood. Normal NMDA receptor (NMDAR) function is essential for neuronal development and higher brain functionality, while its inappropriate stimulation results in neurological deficits. Thus, the current study aimed to investigate the role of NMDARs in CPF-induced neurotoxicity. We show that NMDARs mediate CPF-induced excitotoxicity in differentiated human fetal cortical neuronal ReNcell CX stem cells. In addition, by using two-electrode voltage clamp electrophysiology of Xenopus oocytes expressing NMDARs, we show CPF potentiation of both GluN1-1a/GluN2A (EC50 ≈ 40 nM) and GluN1-1a/GluN2B (EC50 ≈ 55 nM) receptors, as well as reductions (approximately halved) in the NMDA EC50s and direct activation by 10 μM CPF of both receptor types. In silico molecular docking validated CPF's association with NMDARs through relatively high affinity binding (-8.82 kcal/mol) to a modulator site at the GluN1-GluN2A interface of the ligand-binding domains.
Collapse
Affiliation(s)
- Mahmoud Awad Sherif
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Wayne G. Carter
- Clinical Toxicology Research Group, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK;
| | - Ian R. Mellor
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| |
Collapse
|
2
|
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
|
3
|
Sasaki T, Hisada S, Kanki H, Nunomura K, Lin B, Nishiyama K, Kawano T, Matsumura S, Mochizuki H. Modulation of Ca 2+ oscillation following ischemia and nicotinic acetylcholine receptors in primary cortical neurons by high-throughput analysis. Sci Rep 2024; 14:27667. [PMID: 39532929 PMCID: PMC11557898 DOI: 10.1038/s41598-024-77882-w] [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: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Calcium oscillations in primary neuronal cultures and iPSCs have been employed to investigate arrhythmogenicity and epileptogenicity in drug development. Previous studies have demonstrated that Ca2+ influx via NMDA and nicotinic acetylcholine receptors (nAChRs) modulates Ca2+ oscillations. Nevertheless, there has been no comprehensive investigation into the impact of ischemia or nAChR-positive allosteric modulators (PAM) drugs on Ca2+ oscillations at a level that would facilitate high-throughput screening. We investigated the effects of ischemia and nAChR subtypes or nAChR PAM agonists on Ca2+ oscillations in high-density 2D and 3D-sphere primary neuronal cultures using 384-well plates with FDSS-7000. Ischemia for 1 and 2 h resulted in an increase in the frequency of Ca2+ oscillations and a decrease in their amplitude in a time-dependent manner. The NMDA and AMPA receptor inhibition significantly suppressed Ca2+ oscillation. Inhibition of NR2A or NR2B had the opposite effect on Ca oscillations. The potentiation of ischemia-induced Ca2+ oscillations was significantly inhibited by the NMDA receptor antagonist, MK-801, and the frequency of these oscillations was suppressed by the NR2B inhibitor, Ro-256981. In the 3D-neurosphere, the application of an α7nAChR agonist increased the frequency of Ca2+ oscillations, whereas the activation of α4β2 had no effect. The combination of nicotine and PNU-120596 (type II PAM) affected the frequency and amplitude of Ca2+ oscillations in a manner distinct from that of type I PAM. These systems may be useful not only for detecting epileptogenicity but also in the search for neuroprotective agents against cerebral ischemia.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
- StemRIM Institute of Regeneration-Inducing Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| | - Sunao Hisada
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Hideaki Kanki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, 1‑6 Yamadaoka, Suita, Osaka, 565‑0871, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, 1‑6 Yamadaoka, Suita, Osaka, 565‑0871, Japan
| | - Kumiko Nishiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Tomohito Kawano
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Shigenobu Matsumura
- Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, 583-8555, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
4
|
Pedrão LFAT, Medeiros POS, Leandro EC, Falquetto B. Parkinson's disease models and death signaling: what do we know until now? Front Neuroanat 2024; 18:1419108. [PMID: 39533977 PMCID: PMC11555652 DOI: 10.3389/fnana.2024.1419108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/04/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease (PD) is the second neurodegenerative disorder most prevalent in the world, characterized by the loss of dopaminergic neurons in the Substantia Nigra (SN). It is well known for its motor and non-motor symptoms including bradykinesia, resting tremor, psychiatric, cardiorespiratory, and other dysfunctions. Pathological apoptosis contributes to a wide variety of diseases including PD. Various insults and/or cellular phenotypes have been shown to trigger distinct signaling events leading to cell death in neurons affected by PD. The intrinsic or mitochondrial pathway, inflammatory or oxidative stress-induced extrinsic pathways are the main events associated with apoptosis in PD-related neuronal loss. Although SN is the main brain area studied so far, other brain nuclei are also affected by the disease leading to non-classical motor symptoms as well as non-motor symptoms. Among these, the respiratory symptoms are often overlooked, yet they can cause discomfort and may contribute to patients shortened lifespan after disease diagnosis. While animal and in vitro models are frequently used to investigate the mechanisms involved in the pathogenesis of PD in both the SN and other brain regions, these models provide only a limited understanding of the disease's actual progression. This review offers a comprehensive overview of some of the most studied forms of cell death, including recent research on potential treatment targets for these pathways. It highlights key findings and milestones in the field, shedding light on the potential role of understanding cell death in the prevention and treatment of the PD. Therefore, unraveling the connection between these pathways and the notable pathological mechanisms observed during PD progression could enhance our comprehension of the disease's origin and provide valuable insights into potential molecular targets for the developing therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciências Biomédica, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
5
|
Cheng JL, Cook AL, Talbot J, Perry S. How is Excitotoxicity Being Modelled in iPSC-Derived Neurons? Neurotox Res 2024; 42:43. [PMID: 39405005 PMCID: PMC11480214 DOI: 10.1007/s12640-024-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Excitotoxicity linked either to environmental causes (pesticide and cyanotoxin exposure), excitatory neurotransmitter imbalance, or to intrinsic neuronal hyperexcitability, is a pathological mechanism central to neurodegeneration in amyotrophic lateral sclerosis (ALS). Investigation of excitotoxic mechanisms using in vitro and in vivo animal models has been central to understanding ALS mechanisms of disease. In particular, advances in induced pluripotent stem cell (iPSC) technologies now provide human cell-based models that are readily amenable to environmental and network-based excitotoxic manipulations. The cell-type specific differentiation of iPSC, combined with approaches to modelling excitotoxicity that include editing of disease-associated gene variants, chemogenetics, and environmental risk-associated exposures make iPSC primed to examine gene-environment interactions and disease-associated excitotoxic mechanisms. Critical to this is knowledge of which neurotransmitter receptor subunits are expressed by iPSC-derived neuronal cultures being studied, how their activity responds to antagonists and agonists of these receptors, and how to interpret data derived from multi-parameter electrophysiological recordings. This review explores how iPSC-based studies have contributed to our understanding of ALS-linked excitotoxicity and highlights novel approaches to inducing excitotoxicity in iPSC-derived neurons to further our understanding of its pathological pathways.
Collapse
Affiliation(s)
- Jan L Cheng
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia.
| |
Collapse
|
6
|
Dando O, McQueen J, Burr K, Kind PC, Chandran S, Hardingham GE, Qiu J. A comparison of basal and activity-dependent exon splicing in cortical-patterned neurons of human and mouse origin. Front Mol Neurosci 2024; 17:1392408. [PMID: 39268251 PMCID: PMC11390650 DOI: 10.3389/fnmol.2024.1392408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
Rodent studies have shown that alternative splicing in neurons plays important roles in development and maturity, and is regulatable by signals such as electrical activity. However, rodent-human similarities are less well explored. We compared basal and activity-dependent exon splicing in cortical-patterned human ESC-derived neurons with that in cortical mouse ESC-derived neurons, primary mouse cortical neurons at two developmental stages, and mouse hippocampal neurons, focussing on conserved orthologous exons. Both basal exon inclusion levels and activity-dependent changes in splicing showed human-mouse correlation. Conserved activity regulated exons are enriched in RBFOX, SAM68, NOVA and PTBP targets, and centered on cytoskeletal organization, mRNA processing, and synaptic signaling genes. However, human-mouse correlations were weaker than inter-mouse comparisons of neurons from different brain regions, developmental stages and origin (ESC vs. primary), suggestive of some inter-species divergence. The set of genes where activity-dependent splicing was observed only in human neurons were dominated by those involved in lipid biosynthesis, signaling and trafficking. Study of human exon splicing in mouse Tc1 neurons carrying human chromosome-21 showed that neuronal basal exon inclusion was influenced by cis-acting sequences, although may not be sufficient to confer activity-responsiveness in an allospecific environment. Overall, these comparisons suggest that neuronal alternative splicing should be confirmed in a human-relevant system even when exon structure is evolutionarily conserved.
Collapse
Affiliation(s)
- Owen Dando
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie McQueen
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| | - Karen Burr
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter C Kind
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences, Edinburgh Medical School, Edinburgh, United Kingdom
| | - Giles E Hardingham
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| | - Jing Qiu
- Edinburgh Medical School, UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
7
|
Salerno JA, Rehen S. Human pluripotent stem cells as a translational toolkit in psychedelic research in vitro. iScience 2024; 27:109631. [PMID: 38628967 PMCID: PMC11019282 DOI: 10.1016/j.isci.2024.109631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
Psychedelics, recognized for their impact on perception, are resurging as promising treatments with rapid onset for mood and substance use disorders. Despite increasing evidence from clinical trials, questions persist about the cellular and molecular mechanisms and their precise correlation with treatment outcomes. Murine neurons and immortalized non-neural cell lines harboring overexpressed constructs have shed light on neuroplastic changes mediated by the serotonin 2A receptor (5-HT2AR) as the primary mechanism. However, limitations exist in capturing human- and disease-specific traits. Here, we discuss current accomplishments and prospects for incorporating human pluripotent stem cells (PSCs) to complement these models. PSCs can differentiate into various brain cell types, mirroring endogenous expression patterns and cell identities to recreate disease phenotypes. Brain organoids derived from PSCs resemble cell diversity and patterning, while region-specific organoids simulate circuit-level phenotypes. PSC-based models hold significant promise to illuminate the cellular and molecular substrates of psychedelic-induced phenotypic recovery in neuropsychiatric disorders.
Collapse
Affiliation(s)
- José Alexandre Salerno
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Graduate Program in Morphological Sciences, Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Department of Morphological Sciences, Biomedical Institute, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Stevens Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Usona Institute, Fitchburg, WI, USA
- Promega Corporation, Madison, WI, USA
| |
Collapse
|
8
|
Yan L, Li Z, Li C, Chen J, Zhou X, Cui J, Liu P, Shen C, Chen C, Hong H, Xu G, Cui Z. Hspb1 and Lgals3 in spinal neurons are closely associated with autophagy following excitotoxicity based on machine learning algorithms. PLoS One 2024; 19:e0303235. [PMID: 38728287 PMCID: PMC11086895 DOI: 10.1371/journal.pone.0303235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
Excitotoxicity represents the primary cause of neuronal death following spinal cord injury (SCI). While autophagy plays a critical and intricate role in SCI, the specific mechanism underlying the relationship between excitotoxicity and autophagy in SCI has been largely overlooked. In this study, we isolated primary spinal cord neurons from neonatal rats and induced excitotoxic neuronal injury by high concentrations of glutamic acid, mimicking an excitotoxic injury model. Subsequently, we performed transcriptome sequencing. Leveraging machine learning algorithms, including weighted correlation network analysis (WGCNA), random forest analysis (RF), and least absolute shrinkage and selection operator analysis (LASSO), we conducted a comprehensive investigation into key genes associated with spinal cord neuron injury. We also utilized protein-protein interaction network (PPI) analysis to identify pivotal proteins regulating key gene expression and analyzed key genes from public datasets (GSE2599, GSE20907, GSE45006, and GSE174549). Our findings revealed that six genes-Anxa2, S100a10, Ccng1, Timp1, Hspb1, and Lgals3-were significantly upregulated not only in vitro in neurons subjected to excitotoxic injury but also in rats with subacute SCI. Furthermore, Hspb1 and Lgals3 were closely linked to neuronal autophagy induced by excitotoxicity. Our findings contribute to a better understanding of excitotoxicity and autophagy, offering potential targets and a theoretical foundation for SCI diagnosis and treatment.
Collapse
Affiliation(s)
- Lei Yan
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zihao Li
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Chuanbo Li
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jingyu Chen
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xun Zhou
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaming Cui
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Peng Liu
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Chong Shen
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Chu Chen
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Hongxiang Hong
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Guanhua Xu
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhiming Cui
- The First People’s Hospital of Nantong, Research Institute for Spine and Spinal Cord Disease of Nantong University, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
9
|
Starr A, Nickoloff-Bybel E, Abedalthaqafi R, Albloushi N, Jordan-Sciutto KL. Human iPSC-derived neurons reveal NMDAR-independent dysfunction following HIV-associated insults. Front Mol Neurosci 2024; 16:1353562. [PMID: 38348237 PMCID: PMC10859444 DOI: 10.3389/fnmol.2023.1353562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 12/30/2023] [Indexed: 02/15/2024] Open
Abstract
The central nervous system encounters a number of challenges following HIV infection, leading to increased risk for a collection of neurocognitive symptoms clinically classified as HIV-associated neurocognitive disorders (HAND). Studies attempting to identify causal mechanisms and potential therapeutic interventions have historically relied on primary rodent neurons, but a number of recent reports take advantage of iPSC-derived neurons in order to study these mechanisms in a readily reproducible, human model. We found that iPSC-derived neurons differentiated via an inducible neurogenin-2 transcription factor were resistant to gross toxicity from a number of HIV-associated insults previously reported to be toxic in rodent models, including HIV-infected myeloid cell supernatants and the integrase inhibitor antiretroviral drug, elvitegravir. Further examination of these cultures revealed robust resistance to NMDA receptor-mediated toxicity. We then performed a comparative analysis of iPSC neurons exposed to integrase inhibitors and activated microglial supernatants to study sub-cytotoxic alterations in micro electrode array (MEA)-measured neuronal activity and gene expression, identifying extracellular matrix interaction/morphogenesis as the most consistently altered pathways across HIV-associated insults. These findings illustrate that HIV-associated insults dysregulate human neuronal activity and organization even in the absence of gross NMDA-mediated neurotoxicity, which has important implications on the effects of these insults in neurodevelopment and on the interpretation of primary vs. iPSC in vitro neuronal studies.
Collapse
Affiliation(s)
| | | | | | | | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Babkina I, Savinkova I, Molchanova T, Sidorova M, Surin A, Gorbacheva L. Neuroprotective Effects of Noncanonical PAR1 Agonists on Cultured Neurons in Excitotoxicity. Int J Mol Sci 2024; 25:1221. [PMID: 38279219 PMCID: PMC10816171 DOI: 10.3390/ijms25021221] [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: 12/27/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Serine proteases regulate cell functions through G protein-coupled protease-activated receptors (PARs). Cleavage of one peptide bond of the receptor amino terminus results in the formation of a new N-terminus ("tethered ligand") that can specifically interact with the second extracellular loop of the PAR receptor and activate it. Activation of PAR1 by thrombin (canonical agonist) and activated protein C (APC, noncanonical agonist) was described as a biased agonism. Here, we have supposed that synthetic peptide analogs to the PAR1 tethered ligand liberated by APC could have neuroprotective effects like APC. To verify this hypothesis, a model of the ischemic brain impairment based on glutamate (Glu) excitotoxicity in primary neuronal cultures of neonatal rats has been used. It was shown that the nanopeptide NPNDKYEPF-NH2 (AP9) effectively reduced the neuronal death induced by Glu. The influence of AP9 on cell survival was comparable to that of APC. Both APC and AP9 reduced the dysregulation of intracellular calcium homeostasis in cultured neurons induced by excitotoxic Glu (100 µM) or NMDA (200 µM) concentrations. PAR1 agonist synthetic peptides might be noncanonical PAR1 agonists and a basis for novel neuroprotective drugs for disorders related to Glu excitotoxicity such as brain ischemia, trauma and some neurodegenerative diseases.
Collapse
Affiliation(s)
- Irina Babkina
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
| | - Irina Savinkova
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
| | - Tatiana Molchanova
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Maria Sidorova
- Chazov National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia;
| | - Alexander Surin
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Liubov Gorbacheva
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| |
Collapse
|
11
|
Kukułowicz J, Pietrzak-Lichwa K, Klimończyk K, Idlin N, Bajda M. The SLC6A15-SLC6A20 Neutral Amino Acid Transporter Subfamily: Functions, Diseases, and Their Therapeutic Relevance. Pharmacol Rev 2023; 76:142-193. [PMID: 37940347 DOI: 10.1124/pharmrev.123.000886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/07/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.
Collapse
Affiliation(s)
- Jędrzej Kukułowicz
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Pietrzak-Lichwa
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Klimończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Nathalie Idlin
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
12
|
Teng L, Qin Q, Zhou Z, Zhou F, Cao C, Yang J, Ding J. Glutamate secretion by embryonic stem cells as an autocrine signal to promote proliferation. Sci Rep 2023; 13:19069. [PMID: 37925518 PMCID: PMC10625544 DOI: 10.1038/s41598-023-46477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023] Open
Abstract
Glutamate, the major excitatory neurotransmitter in the central nervous system, has also been found to play a role in embryonic stem (ES) cells. However, the exact mechanism and function of glutamatergic signaling in ES cells remain poorly understood. In this study, we identified a glutamatergic transmission circuit in ES cells that operates through an autocrine mechanism and regulates cell proliferation. We performed biological analyses to identify the key components involved in glutamate biosynthesis, packaging for secretion, reaction, and reuptake in ES cells, including glutaminase, vesicular glutamate transporter, glutamate N-methyl-D-aspartate (NMDA) receptor, and cell membrane excitatory amino-acid transporter (EAAT). We directly quantified the released glutamate signal using microdialysis-high performance liquid chromatography-tandem mass spectrometry (MD-HPLC-MS-MS). Pharmacological inhibition of endogenous glutamate release and the resulting tonic activation of NMDA receptors significantly affected ES cell proliferation, suggesting that ES cells establish a glutamatergic autocrine niche via releasing and responding to the transmitter for their own regulation.
Collapse
Affiliation(s)
- Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Ziyi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Chunyu Cao
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Jian Yang
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Jiawang Ding
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China.
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China.
| |
Collapse
|
13
|
Protective Effect of Resveratrol in an Experimental Model of Salicylate-Induced Tinnitus. Int J Mol Sci 2022; 23:ijms232214183. [PMID: 36430660 PMCID: PMC9692321 DOI: 10.3390/ijms232214183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
To date, the effect of resveratrol on tinnitus has not been reported. The attenuative effects of resveratrol (RSV) on a salicylate-induced tinnitus model were evaluated by in vitro and in vivo experiments. The gene expression of the activity-regulated cytoskeleton-associated protein (ARC), tumor necrosis factor-alpha (TNFα), and NMDA receptor subunit 2B (NR2B) in SH-SY5Y cells was examined using qPCR. Phosphorylated cAMP response element-binding protein (p-CREB), apoptosis markers, and reactive oxygen species (ROS) were evaluated by in vitro experiments. The in vivo experiment evaluated the gap-prepulse inhibition of the acoustic startle reflex (GPIAS) and auditory brainstem response (ABR) level. The NR2B expression in the auditory cortex (AC) was determined by immunohistochemistry. RSV significantly reduced the salicylate-induced expression of NR2B, ARC, and TNFα in neuronal cells; the GPIAS and ABR thresholds altered by salicylate in rats were recovered close to their normal range. RSV also reduced the salicylate-induced NR2B overexpression of the AC. These results confirmed that resveratrol exerted an attenuative effect on salicylate-induced tinnitus and may have a therapeutic potential.
Collapse
|
14
|
Haddow K, Kind PC, Hardingham GE. NMDA Receptor C-Terminal Domain Signalling in Development, Maturity, and Disease. Int J Mol Sci 2022; 23:ijms231911392. [PMID: 36232696 PMCID: PMC9570437 DOI: 10.3390/ijms231911392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
The NMDA receptor is a Ca2+-permeant glutamate receptor which plays key roles in health and disease. Canonical NMDARs contain two GluN2 subunits, of which 2A and 2B are predominant in the forebrain. Moreover, the relative contribution of 2A vs. 2B is controlled both developmentally and in an activity-dependent manner. The GluN2 subtype influences the biophysical properties of the receptor through difference in their N-terminal extracellular domain and transmembrane regions, but they also have large cytoplasmic Carboxyl (C)-terminal domains (CTDs) which have diverged substantially during evolution. While the CTD identity does not influence NMDAR subunit specific channel properties, it determines the nature of CTD-associated signalling molecules and has been implicated in mediating the control of subunit composition (2A vs. 2B) at the synapse. Historically, much of the research into the differential function of GluN2 CTDs has been conducted in vitro by over-expressing mutant subunits, but more recently, the generation of knock-in (KI) mouse models have allowed CTD function to be probed in vivo and in ex vivo systems without heterologous expression of GluN2 mutants. In some instances, findings involving KI mice have been in disagreement with models that were proposed based on earlier approaches. This review will examine the current research with the aim of addressing these controversies and how methodology may contribute to differences between studies. We will also discuss the outstanding questions regarding the role of GluN2 CTD sequences in regulating NMDAR subunit composition, as well as their relevance to neurodegenerative disease and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kirsty Haddow
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Peter C. Kind
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Giles E. Hardingham
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Correspondence:
| |
Collapse
|
15
|
AL-Nasser MN, Mellor IR, Carter WG. Is L-Glutamate Toxic to Neurons and Thereby Contributes to Neuronal Loss and Neurodegeneration? A Systematic Review. Brain Sci 2022; 12:577. [PMID: 35624964 PMCID: PMC9139234 DOI: 10.3390/brainsci12050577] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
L-glutamate (L-Glu) is a nonessential amino acid, but an extensively utilised excitatory neurotransmitter with critical roles in normal brain function. Aberrant accumulation of L-Glu has been linked to neurotoxicity and neurodegeneration. To investigate this further, we systematically reviewed the literature to evaluate the effects of L-Glu on neuronal viability linked to the pathogenesis and/or progression of neurodegenerative diseases (NDDs). A search in PubMed, Medline, Embase, and Web of Science Core Collection was conducted to retrieve studies that investigated an association between L-Glu and pathology for five NDDs: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Together, 4060 studies were identified, of which 71 met eligibility criteria. Despite several inadequacies, including small sample size, employment of supraphysiological concentrations, and a range of administration routes, it was concluded that exposure to L-Glu in vitro or in vivo has multiple pathogenic mechanisms that influence neuronal viability. These mechanisms include oxidative stress, reduced antioxidant defence, neuroinflammation, altered neurotransmitter levels, protein accumulations, excitotoxicity, mitochondrial dysfunction, intracellular calcium level changes, and effects on neuronal histology, cognitive function, and animal behaviour. This implies that clinical and epidemiological studies are required to assess the potential neuronal harm arising from excessive intake of exogenous L-Glu.
Collapse
Affiliation(s)
- Maryam N. AL-Nasser
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| | - Ian R. Mellor
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Wayne G. Carter
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| |
Collapse
|
16
|
Kaplan-Arabaci O, Acari A, Ciftci P, Gozuacik D. Glutamate Scavenging as a Neuroreparative Strategy in Ischemic Stroke. Front Pharmacol 2022; 13:866738. [PMID: 35401202 PMCID: PMC8984161 DOI: 10.3389/fphar.2022.866738] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/08/2022] [Indexed: 12/17/2022] Open
Abstract
Stroke is the second highest reason of death in the world and the leading cause of disability. The ischemic stroke makes up the majority of stroke cases that occur due to the blockage of blood vessels. Therapeutic applications for ischemic stroke include thrombolytic treatments that are in limited usage and only applicable to less than 10% of the total stroke patients, but there are promising new approaches. The main cause of ischemic neuronal death is glutamate excitotoxicity. There have been multiple studies focusing on neuroprotection via reduction of glutamate both in ischemic stroke and other neurodegenerative diseases that ultimately failed due to the obstacles in delivery. At that point, systemic glutamate grabbing, or scavenging is an ingenious way of decreasing glutamate levels upon ischemic stroke. The main advantage of this new therapeutic method is the scavengers working in the circulating blood so that there is no interference with the natural brain neurophysiology. In this review, we explain the molecular mechanisms of ischemic stroke, provide brief information about existing drugs and approaches, and present novel systemic glutamate scavenging methods. This review hopefully will elucidate the potential usage of the introduced therapeutic approaches in stroke patients.
Collapse
Affiliation(s)
- Oykum Kaplan-Arabaci
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Alperen Acari
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Pinar Ciftci
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey.,School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
17
|
Kang JB, Park DJ, Shah MA, Koh PO. Quercetin ameliorates glutamate toxicity-induced neuronal cell death by controlling calcium-binding protein parvalbumin. J Vet Sci 2022; 23:e26. [PMID: 35187882 PMCID: PMC8977545 DOI: 10.4142/jvs.21273] [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: 10/20/2021] [Revised: 12/06/2021] [Accepted: 12/23/2021] [Indexed: 11/24/2022] Open
Abstract
Background Glutamate is the main excitatory neurotransmitter. Excessive glutamate causes excitatory toxicity and increases intracellular calcium, leading to neuronal death. Parvalbumin is a calcium-binding protein that regulates calcium homeostasis. Quercetin is a polyphenol found in plant and has neuroprotective effects against neurodegenerative diseases. Objectives We investigated whether quercetin regulates apoptosis by modulating parvalbumin expression in glutamate induced neuronal damage. Methods Glutamate was treated in hippocampal-derived cell line, and quercetin or vehicle was treated 1 h before glutamate exposure. Cells were collected for experimental procedure 24 h after glutamate treatment and intracellular calcium concentration and parvalbumin expression were examined. Parvalbumin small interfering RNA (siRNA) transfection was performed to detect the relation between parvalbumin and apoptosis. Results Glutamate reduced cell viability and increased intracellular calcium concentration, while quercetin preserved calcium concentration and neuronal damage. Moreover, glutamate reduced parvalbumin expression and quercetin alleviated this reduction. Glutamate increased caspase-3 expression, and quercetin attenuated this increase in both parvalbumin siRNA transfected and non-transfected cells. The alleviative effect of quercetin was statistically significant in non-transfected cells. Moreover, glutamate decreased bcl-2 and increased bax expressions, while quercetin alleviated these changes. The alleviative effect of quercetin in bcl-2 family protein expression was more remarkable in non-transfected cells. Conclusions These results demonstrate that parvalbumin contributes to the maintainace of intracellular calcium concentration and the prevention of apoptosis, and quercetin modulates parvalbumin expression in glutamate-exposed cells. Thus, these findings suggest that quercetin performs neuroprotective function against glutamate toxicity by regulating parvalbumin expression.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Dong-Ju Park
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Murad-Ali Shah
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Phil-Ok Koh
- Department of Anatomy and Histology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
18
|
Bauersachs HG, Bengtson CP, Weiss U, Hellwig A, García-Vilela C, Zaremba B, Kaessmann H, Pruunsild P, Bading H. N-methyl-d-aspartate Receptor-mediated Preconditioning Mitigates Excitotoxicity in Human induced Pluripotent Stem Cell-derived Brain Organoids. Neuroscience 2021; 484:83-97. [DOI: 10.1016/j.neuroscience.2021.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/15/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022]
|
19
|
Al-Mubarak BR, Bell KFS, Chowdhry S, Meakin PJ, Baxter PS, McKay S, Dando O, Ashford MLJ, Gazaryan I, Hayes JD, Hardingham GE. Non-canonical Keap1-independent activation of Nrf2 in astrocytes by mild oxidative stress. Redox Biol 2021; 47:102158. [PMID: 34626892 PMCID: PMC8512624 DOI: 10.1016/j.redox.2021.102158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Nrf2 is a stress-responsive master regulator of antioxidant, detoxification and proteostasis genes. In astrocytes, Nrf2-dependent gene expression drives cell-autonomous cytoprotection and also non-cell-autonomous protection of nearby neurons, and can ameliorate pathology in several acute and chronic neurological disorders associated with oxidative stress. However, the value of astrocytic Nrf2 as a therapeutic target depends in part on whether Nrf2 activation by disease-associated oxidative stress occludes the effect of any Nrf2-activating drug. Nrf2 activation classically involves the inhibition of interactions between Nrf2's Neh2 domain and Keap1, which directs Nrf2 degradation. Keap1 inhibition is mediated by the modification of cysteine residues on Keap1, and can be triggered by electrophilic small molecules such as tBHQ. Here we show that astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling. Keap1 deficiency elevates basal Nrf2 target gene expression in astrocytes and occludes the effects of tBHQ, oxidative stress still induced strong Nrf2-dependent gene expression in Keap1-deficient astrocytes. Moreover, while tBHQ prevented protein degradation mediated via Nrf2's Neh2 domain, oxidative stress did not, consistent with a Keap1-independent mechanism. Moreover the effects of oxidative stress and tBHQ on Nrf2 target gene expression are additive, not occlusive. Mechanistically, oxidative stress enhances the transactivation potential of Nrf2's Neh5 domain in a manner dependent on its Cys-191 residue. Thus, astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling, meaning that further Nrf2 activation by Keap1-inhibiting drugs may be a viable therapeutic strategy.
Collapse
Affiliation(s)
- Bashayer R Al-Mubarak
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; Behavioral Genetics Unit, Department of Genetics, King Faisal Specialist Hospital and Research Center, P.O Box 3354, Riyadh, 11211, Saudi Arabia
| | - Karen F S Bell
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Sudhir Chowdhry
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Paul J Meakin
- Discovery & Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, LS2 9JT, UK; Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Paul S Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Sean McKay
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK
| | - Michael L J Ashford
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Irina Gazaryan
- Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY, 10570, USA
| | - John D Hayes
- Biomedical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, EH16 4SB, UK.
| |
Collapse
|
20
|
D’Ezio V, Colasanti M, Persichini T. Amyloid-β 25-35 Induces Neurotoxicity through the Up-Regulation of Astrocytic System X c. Antioxidants (Basel) 2021;10:antiox10111685. [PMID: 34829555 PMCID: PMC8615014 DOI: 10.3390/antiox10111685] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022] Open
Abstract
Amyloid-β (Aβ) deposition, a hallmark of Alzheimer’s disease, is known to induce free radical production and oxidative stress, leading to neuronal damage. During oxidative stress, several cell types (including astrocytes) can activate the nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of several phase II detoxifying and antioxidant genes, such as the System Xc− subunit xCT. Here, we studied (i) the effect of the Aβ fragment 25-35 (Aβ25-35) on Nrf2-dependent System Xc− expression in U373 human astroglial cells and (ii) the effect of Aβ25-35-induced astrocytic response on neuronal cell viability using an in vitro co-culture system. We found that Aβ25-35 was able to activate an antioxidant response in astrocytes, by inducing both Nrf2 activation and System Xc− up-regulation. However, this astrocytic response caused an enhanced cell mortality of co-cultured SH-SY5Y cells, taken as a neuronal model. Consistently, the specific System Xc− inhibitor sulfasalazine prevented the increase of both neuronal mortality and extracellular glutamate levels, thus indicating that the neurotoxic effect was due to an augmented release of glutamate through the transporter. The involvement of NMDA receptor activation in this pathway was also demonstrated using the specific inhibitor MK801 that completely restored neuronal viability at the control levels. The present study sheds light on the Nrf2/system Xc− pathway in the toxicity induced by Aβ25-35 and may help to better understand the involvement of astrocytes in neuronal death during Alzheimer’s disease.
Collapse
|
21
|
Barua S, Sim AY, Kim JY, Shin I, Lee JE. Maintenance of the Neuroprotective Function of the Amino Group Blocked Fluorescence-Agmatine. Neurochem Res 2021; 46:1933-1940. [PMID: 33914233 PMCID: PMC8254702 DOI: 10.1007/s11064-021-03319-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/21/2021] [Accepted: 04/02/2021] [Indexed: 11/23/2022]
Abstract
Agmatine, an endogenous derivative of arginine, has been found to be effective in treating idiopathic pain, convulsion, stress-mediated behavior, and attenuate the withdrawal symptoms of drugs like morphine. In the early stages of ischemic brain injury in animals, exogenous agmatine treatment was found to be neuroprotective. Agmatine is also considered as a putative neurotransmitter and is still an experimental drug. Chemically, agmatine is called agmatine 1-(4-aminobutyl guanidine). Crystallographic study data show that positively-charged guanidine can bind to the protein containing Gly and Asp residues, and the amino group can interact with the complimentary sites of Glu and Ser. In this study, we blocked the amino end of the agmatine by conjugating it with FITC, but the guanidine end was unchanged. We compared the neuroprotective function of the agmatine and agmatine-FITC by treating them in neurons after excitotoxic stimulation. We found that even the amino end blocked neuronal viability in the excitotoxic condition, by NMDA treatment for 1 h, was increased by agmatine-FITC, which was similar to that of agmatine. We also found that the agmatine-FITC treatment reduced the expression of nitric oxide production in NMDA-treated cells. This study suggests that even if the amino end of agmatine is blocked, it can perform its neuroprotective function.
Collapse
Affiliation(s)
- Sumit Barua
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 03722 Korea
| | - A Young Sim
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 03722 Korea
- BK21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722 Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 03722 Korea
| | - Injae Shin
- Department of Chemistry, Yonsei University, Seoul, 03722 Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul, 03722 Korea
- BK21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722 Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, 03722 Korea
| |
Collapse
|
22
|
Shahsavani N, Alizadeh A, Kataria H, Karimi-Abdolrezaee S. Availability of neuregulin-1beta1 protects neurons in spinal cord injury and against glutamate toxicity through caspase dependent and independent mechanisms. Exp Neurol 2021; 345:113817. [PMID: 34314724 DOI: 10.1016/j.expneurol.2021.113817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/06/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) causes sensorimotor and autonomic impairment that partly reflects extensive, permanent loss of neurons at the epicenter and penumbra of the injury. Strategies aimed at enhancing neuronal protection are critical to attenuate neurodegeneration and improve neurological recovery after SCI. In rat SCI, we previously uncovered that the tissue levels of neuregulin-1beta 1 (Nrg-1β1) are acutely and persistently downregulated in the injured spinal cord. Nrg-1β1 is well-known for its critical roles in the development, maintenance and physiology of neurons and glia in the developing and adult spinal cord. However, despite this pivotal role, Nrg-1β1 specific effects and mechanisms of action on neuronal injury remain largely unknown in SCI. In the present study, using a clinically-relevant model of compressive/contusive SCI in rats and an in vitro model of glutamate toxicity in primary neurons, we demonstrate Nrg-1β1 provides early neuroprotection through attenuation of reactive oxygen species, lipid peroxidation, necrosis and apoptosis in acute and subacute stages of SCI. Mechanistically, availability of Nrg-1β1 following glutamate challenge protects neurons from caspase-dependent and independent cell death that is mediated by modulation of mitochondria associated apoptotic cascades and MAP kinase and AKT signaling pathways. Altogether, our work provides novel insights into the role and mechanisms of Nrg-1β1 in neuronal injury after SCI and introduces its potential as a new neuroprotective target for this debilitating neurological condition.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
23
|
Bridging Cyanobacteria to Neurodegenerative Diseases: A New Potential Source of Bioactive Compounds against Alzheimer's Disease. Mar Drugs 2021; 19:md19060343. [PMID: 34208482 PMCID: PMC8235772 DOI: 10.3390/md19060343] [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] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 02/02/2023] Open
Abstract
Neurodegenerative diseases (NDs) represent a drawback in society given the ageing population. Dementias are the most prevalent NDs, with Alzheimer’s disease (AD) representing around 70% of all cases. The current pharmaceuticals for AD are symptomatic and with no effects on the progression of the disease. Thus, research on molecules with therapeutic relevance has become a major focus for the scientific community. Cyanobacteria are a group of photosynthetic prokaryotes rich in biomolecules with confirmed activity in pathologies such as cancer, and with feasible potential in NDs such as AD. In this review, we aimed to compile the research works focused in the anti-AD potential of cyanobacteria, namely regarding the inhibition of the enzyme β-secretase (BACE1) as a fundamental enzyme in the generation of β-amyloid (Aβ), the inhibition of the enzyme acetylcholinesterase (AChE) lead to an increase in the availability of the neurotransmitter acetylcholine in the synaptic cleft and the antioxidant and anti-inflammatory effects, as phenomena associated with neurodegeneration mechanisms.
Collapse
|
24
|
Naldan ME, Taghizadehghalehjoughi A. Remifentanil reduces glutamate toxicity in rat olfactory bulb neurons in culture. Braz J Anesthesiol 2021; 71:402-407. [PMID: 33895216 PMCID: PMC9373102 DOI: 10.1016/j.bjane.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 11/28/2022] Open
Abstract
Background Opioids are widely used as an analgesic drug in the surgical setting. Remifentanil is an ultra-short acting opioid with selective affinity to the mu (μ) receptor, and also exhibits GABA agonist effects. The aim of this study was study of the neurotoxic or neuroprotective effect of different doses of remifentanil in glutamate-induced toxicity in olfactory neuron cell culture. Materials and methods Olfactory neurons were obtained from newborn Sprague Dawley rat pups. Glutamate 10-5 mM was added to all culture dishes, except for the negative control group. Remifentanil was added at three different doses for 24 hours, after which evaluation was performed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Total Antioxidant Capacity (TAC), Total Oxidant Status (TOS), and Annexin V. Results The highest and lowest viability values were obtained from the low and high remifentanil doses at approximately 91% and 75%, respectively. TAC and TOS were correlated with the MTT results. TAC, TOS and MTT most closely approximated to the sham group values in the remifentanil 0.02 mM group. Conclusions Our results suggest that remifentanil has the potential to reduce glutamate toxicity and to increase cell viability in cultured neuron from the rat olfactory bulb.
Collapse
Affiliation(s)
- Muhammet Emin Naldan
- Erzurum Regional Training and Research Hospital, Department of Anesthesiology and Reanimation, Erzurum, Turkey.
| | - Ali Taghizadehghalehjoughi
- Atatürk University, Faculty of Veterinary Science, Department of Pharmacology and Toxicology, Erzurum, Turkey
| |
Collapse
|
25
|
Todd AC, Hardingham GE. The Regulation of Astrocytic Glutamate Transporters in Health and Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E9607. [PMID: 33348528 PMCID: PMC7766851 DOI: 10.3390/ijms21249607] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
The astrocytic glutamate transporters excitatory amino acid transporters 1 and 2 (EAAT1 and EAAT2) play a key role in nervous system function to maintain extracellular glutamate levels at low levels. In physiology, this is essential for the rapid uptake of synaptically released glutamate, maintaining the temporal fidelity of synaptic transmission. However, EAAT1/2 hypo-expression or hypo-function are implicated in several disorders, including epilepsy and neurodegenerative diseases, as well as being observed naturally with aging. This not only disrupts synaptic information transmission, but in extremis leads to extracellular glutamate accumulation and excitotoxicity. A key facet of EAAT1/2 expression in astrocytes is a requirement for signals from other brain cell types in order to maintain their expression. Recent evidence has shown a prominent role for contact-dependent neuron-to-astrocyte and/or endothelial cell-to-astrocyte Notch signalling for inducing and maintaining the expression of these astrocytic glutamate transporters. The relevance of this non-cell-autonomous dependence to age- and neurodegenerative disease-associated decline in astrocytic EAAT expression is discussed, plus the implications for disease progression and putative therapeutic strategies.
Collapse
Affiliation(s)
- Alison C. Todd
- UK Dementia Research Institute at the University of Edinburgh, Chancellor’s Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK;
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Giles E. Hardingham
- UK Dementia Research Institute at the University of Edinburgh, Chancellor’s Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK;
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
26
|
Marino J, Maubert ME, Mele AR, Spector C, Wigdahl B, Nonnemacher MR. Functional impact of HIV-1 Tat on cells of the CNS and its role in HAND. Cell Mol Life Sci 2020; 77:5079-5099. [PMID: 32577796 PMCID: PMC7674201 DOI: 10.1007/s00018-020-03561-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (Tat) is a potent mediator involved in the development of HIV-1-associated neurocognitive disorders (HAND). Tat is expressed even in the presence of antiretroviral therapy (ART) and is able to enter the central nervous system (CNS) through a variety of ways, where Tat can interact with microglia, astrocytes, brain microvascular endothelial cells, and neurons. The presence of low concentrations of extracellular Tat alone has been shown to lead to dysregulated gene expression, chronic cell activation, inflammation, neurotoxicity, and structural damage in the brain. The reported effects of Tat are dependent in part on the specific HIV-1 subtype and amino acid length of Tat used. HIV-1 subtype B Tat is the most common subtype in North American and therefore, most studies have been focused on subtype B Tat; however, studies have shown many genetic, biologic, and pathologic differences between HIV subtype B and subtype C Tat. This review will focus primarily on subtype B Tat where the full-length protein is 101 amino acids, but will also consider variants of Tat, such as Tat 72 and Tat 86, that have been reported to exhibit a number of distinctive activities with respect to mediating CNS damage and neurotoxicity.
Collapse
Affiliation(s)
- Jamie Marino
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Monique E Maubert
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Anthony R Mele
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Cassandra Spector
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Bancroft EA, Srinivasan R. Quantifying Spontaneous Ca2+ Fluxes and their Downstream Effects in Primary Mouse Midbrain Neurons. J Vis Exp 2020. [PMID: 32986023 DOI: 10.3791/61481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a devastating neurodegenerative disorder caused by the degeneration of dopaminergic (DA) neurons. Excessive Ca2+ influx due to the abnormal activation of glutamate receptors results in DA excitotoxicity and has been identified as an important mechanism for DA neuron loss. In this study, we isolate, dissociate, and culture midbrain neurons from the mouse ventral mesencephalon (VM) of ED14 mouse embryos. We then infect the long-term primary mouse midbrain cultures with an adeno-associated virus (AAV) expressing a genetically encoded calcium indicator, GCaMP6f under control of the human neuron-specific synapsin promoter, hSyn. Using live confocal imaging, we show that cultured mouse midbrain neurons display spontaneous Ca2+ fluxes detected by AAV-hSyn-GCaMP6f. Bath application of glutamate to midbrain cultures causes abnormal elevations in intracellular Ca2+ within neurons and this is accompanied by caspase-3 activation in DA neurons, as demonstrated by immunostaining. The techniques to identify glutamate-mediated apoptosis in primary mouse DA neurons have important applications for the high content screening of drugs that preserve DA neuron health.
Collapse
Affiliation(s)
- Eric A Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine; Texas A&M Institute for Neuroscience;
| |
Collapse
|
28
|
Two Faces of Heme Catabolic Pathway in Newborns: A Potential Role of Bilirubin and Carbon Monoxide in Neonatal Inflammatory Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7140496. [PMID: 32908636 PMCID: PMC7450323 DOI: 10.1155/2020/7140496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
In an infant's body, all the systems undergo significant changes in order to adapt to the new, extrauterine environment and challenges which it poses. Fragile homeostasis can be easily disrupted as the defensive mechanisms are yet imperfect. The activity of antioxidant enzymes, i.e., superoxide dismutase, catalase, and glutathione peroxidase, is low; therefore, neonates are especially vulnerable to oxidative stress. Free radical burden significantly contributes to neonatal illnesses such as sepsis, retinopathy of premature, necrotizing enterocolitis, bronchopulmonary dysplasia, or leukomalacia. However, newborns have an important ally-an inducible heme oxygenase-1 (HO-1) which expression rises rapidly in response to stress stimuli. HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Although CO and bilirubin used to be considered noxious by-products, new interesting properties of those compounds are being revealed. Bilirubin proved to be an efficient free radicals scavenger and modulator of immune responses. CO affects a vast range of processes such as vasodilatation, platelet aggregation, and inflammatory reactions. Recently, developed nanoparticles consisting of PEGylated bilirubin as well as several kinds of molecules releasing CO have been successfully tested on animal models of inflammatory diseases. This paper focuses on the role of heme metabolites and their potential utility in prevention and treatment of neonatal diseases.
Collapse
|
29
|
Belov Kirdajova D, Kriska J, Tureckova J, Anderova M. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front Cell Neurosci 2020; 14:51. [PMID: 32265656 PMCID: PMC7098326 DOI: 10.3389/fncel.2020.00051] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
A plethora of neurological disorders shares a final common deadly pathway known as excitotoxicity. Among these disorders, ischemic injury is a prominent cause of death and disability worldwide. Brain ischemia stems from cardiac arrest or stroke, both responsible for insufficient blood supply to the brain parenchyma. Glucose and oxygen deficiency disrupts oxidative phosphorylation, which results in energy depletion and ionic imbalance, followed by cell membrane depolarization, calcium (Ca2+) overload, and extracellular accumulation of excitatory amino acid glutamate. If tight physiological regulation fails to clear the surplus of this neurotransmitter, subsequent prolonged activation of glutamate receptors forms a vicious circle between elevated concentrations of intracellular Ca2+ ions and aberrant glutamate release, aggravating the effect of this ischemic pathway. The activation of downstream Ca2+-dependent enzymes has a catastrophic impact on nervous tissue leading to cell death, accompanied by the formation of free radicals, edema, and inflammation. After decades of “neuron-centric” approaches, recent research has also finally shed some light on the role of glial cells in neurological diseases. It is becoming more and more evident that neurons and glia depend on each other. Neuronal cells, astrocytes, microglia, NG2 glia, and oligodendrocytes all have their roles in what is known as glutamate excitotoxicity. However, who is the main contributor to the ischemic pathway, and who is the unsuspecting victim? In this review article, we summarize the so-far-revealed roles of cells in the central nervous system, with particular attention to glial cells in ischemia-induced glutamate excitotoxicity, its origins, and consequences.
Collapse
Affiliation(s)
- Denisa Belov Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
30
|
Qiu J, Dando O, Febery JA, Fowler JH, Chandran S, Hardingham GE. Neuronal Activity and Its Role in Controlling Antioxidant Genes. Int J Mol Sci 2020; 21:ijms21061933. [PMID: 32178355 PMCID: PMC7139385 DOI: 10.3390/ijms21061933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 12/14/2022] Open
Abstract
Forebrain neurons have relatively weak intrinsic antioxidant defenses compared to astrocytes, in part due to hypo-expression of Nrf2, an oxidative stress-induced master regulator of antioxidant and detoxification genes. Nevertheless, neurons do possess the capacity to auto-regulate their antioxidant defenses in response to electrical activity. Activity-dependent Ca2+ signals control the expression of several antioxidant genes, boosting redox buffering capacity, thus meeting the elevated antioxidant requirements associated with metabolically expensive electrical activity. These genes include examples which are reported Nrf2 target genes and yet are induced in a Nrf2-independent manner. Here we discuss the implications for Nrf2 hypofunction in neurons and the mechanisms underlying the Nrf2-independent induction of antioxidant genes by electrical activity. A significant proportion of Nrf2 target genes, defined as those genes controlled by Nrf2 in astrocytes, are regulated by activity-dependent Ca2+ signals in human stem cell-derived neurons. We propose that neurons interpret Ca2+ signals in a similar way to other cell types sense redox imbalance, to broadly induce antioxidant and detoxification genes.
Collapse
Affiliation(s)
- Jing Qiu
- UK Dementia Research Institute, The Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK; (J.Q.); (O.D.); (S.C.)
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; (J.A.F.); (J.H.F.)
| | - Owen Dando
- UK Dementia Research Institute, The Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK; (J.Q.); (O.D.); (S.C.)
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; (J.A.F.); (J.H.F.)
| | - James A. Febery
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; (J.A.F.); (J.H.F.)
| | - Jill H. Fowler
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; (J.A.F.); (J.H.F.)
| | - Siddharthan Chandran
- UK Dementia Research Institute, The Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK; (J.Q.); (O.D.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh Chancellor’s Building, Edinburgh, EH16 4SB, UK
| | - Giles E. Hardingham
- UK Dementia Research Institute, The Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK; (J.Q.); (O.D.); (S.C.)
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; (J.A.F.); (J.H.F.)
- Correspondence:
| |
Collapse
|
31
|
Szychowski KA, Gmiński J. Specific role of N-methyl-D-aspartate (NMDA) receptor in elastin-derived VGVAPG peptide-dependent calcium homeostasis in mouse cortical astrocytes in vitro. Sci Rep 2019; 9:20165. [PMID: 31882909 PMCID: PMC6934688 DOI: 10.1038/s41598-019-56781-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/10/2019] [Indexed: 12/26/2022] Open
Abstract
Under physiological and pathological conditions, elastin is degraded to produce elastin-derived peptides (EDPs). EDPs are detected in the healthy human brain, and its concentration significantly increases after ischemic stroke. Both elastin and EDPs contains replications of the soluble VGVAPG hexapeptide, which has a broad range of biological activities. Effects of VGVAPG action are mainly mediated by elastin-binding protein (EBP), which is alternatively spliced, enzymatically inactive form of the GLB1 gene. This study was conducted to elucidate the activation and role of the N-methyl-D-aspartate receptor (NMDAR) in elastin-derived VGVAPG peptide-dependent calcium homeostasis in mouse cortical astrocytes in vitro. Cells were exposed to 10 nM VGVAPG peptide and co-treated with MK-801, nifedipine, verapamil, or Src kinase inhibitor I. After cell stimulation, we measured Ca2+ level, ROS production, and mRNA expression. Moreover, the Glb1 and NMDAR subunits (GluN1, GluN2A, and GluN2B) siRNA gene knockdown were applied. We found the VGVAPG peptide causes Ca2+ influx through the NMDA receptor in mouse astrocytes in vitro. Silencing of the Glb1, GluN1, GluN2A, and GluN2B gene prevented VGVAPG peptide-induced increase in Ca2+. Nifedipine does not completely reduce VGVAPG peptide-activated ROS production, whereas MK-801, verapamil, and Src inhibitor reduce VGVAPG peptide-activated Ca2+ influx and ROS production. These data suggest the role of Src kinase signal transduction from EBP to NMDAR. Moreover, the VGVAPG peptide affects the expression of NMDA receptor subunits.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland.
| | - Jan Gmiński
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| |
Collapse
|
32
|
Nehme R, Zuccaro E, Ghosh SD, Li C, Sherwood JL, Pietilainen O, Barrett LE, Limone F, Worringer KA, Kommineni S, Zang Y, Cacchiarelli D, Meissner A, Adolfsson R, Haggarty S, Madison J, Muller M, Arlotta P, Fu Z, Feng G, Eggan K. Combining NGN2 Programming with Developmental Patterning Generates Human Excitatory Neurons with NMDAR-Mediated Synaptic Transmission. Cell Rep 2019; 23:2509-2523. [PMID: 29791859 PMCID: PMC6003669 DOI: 10.1016/j.celrep.2018.04.066] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/07/2018] [Accepted: 04/14/2018] [Indexed: 11/05/2022] Open
Abstract
Transcription factor programming of pluripotent stem cells (PSCs) has emerged as an approach to generate human neurons for disease modeling. However, programming schemes produce a variety of cell types, and those neurons that are made often retain an immature phenotype, which limits their utility in modeling neuronal processes, including synaptic transmission. We report that combining NGN2 programming with SMAD and WNT inhibition generates human patterned induced neurons (hpiNs). Single-cell analyses showed that hpiN cultures contained cells along a developmental continuum, ranging from poorly differentiated neuronal progenitors to well-differentiated, excitatory glutamatergic neurons. The most differentiated neurons could be identified using a CAMK2A::GFP reporter gene and exhibited greater functionality, including NMDAR-mediated synaptic transmission. We conclude that utilizing single-cell and reporter gene approaches for selecting successfully programmed cells for study will greatly enhance the utility of hpiNs and other programmed neuronal populations in the modeling of nervous system disorders.
Collapse
Affiliation(s)
- Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Emanuela Zuccaro
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sulagna Dia Ghosh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Chenchen Li
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - John L Sherwood
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Olli Pietilainen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Lindy E Barrett
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Francesco Limone
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | - Sravya Kommineni
- Novartis Institutes for Biomedical Research, Novartis, Cambridge, MA 02139, USA
| | - Ying Zang
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Davide Cacchiarelli
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alex Meissner
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Rolf Adolfsson
- Umea University, Faculty of Medicine, Department of Clinical Sciences, Psychiatry, 901 85 Umea, Sweden
| | - Stephen Haggarty
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jon Madison
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Matthias Muller
- Novartis Institutes for Biomedical Research, Novartis, 4056 Basel, Switzerland
| | - Paola Arlotta
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; McGovern Institute for Brain Research in the Department of Brain and Cognitive Sciences at MIT, Cambridge, MA 02139, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
33
|
Deb S, Dutta A, Phukan BC, Manivasagam T, Justin Thenmozhi A, Bhattacharya P, Paul R, Borah A. Neuroprotective attributes of L-theanine, a bioactive amino acid of tea, and its potential role in Parkinson's disease therapeutics. Neurochem Int 2019; 129:104478. [DOI: 10.1016/j.neuint.2019.104478] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/17/2019] [Accepted: 05/26/2019] [Indexed: 10/26/2022]
|
34
|
Fjelldal MF, Hadera MG, Kongstorp M, Austdal LPE, Šulović A, Andersen JM, Paulsen RE. Opioid receptor-mediated changes in the NMDA receptor in developing rat and chicken. Int J Dev Neurosci 2019; 78:19-27. [PMID: 31351113 DOI: 10.1016/j.ijdevneu.2019.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/09/2019] [Accepted: 07/23/2019] [Indexed: 11/30/2022] Open
Abstract
The use of opioids during pregnancy has been associated with neurodevelopmental toxicity in exposed children, leading to cognitive and behavioural deficits later in life. The N-methyl-D-aspartate receptor (NMDAR) subunit GluN2B plays critical roles in cerebellar development, and methadone has been shown to possess NMDAR antagonist effect. Consequently, we wanted to explore if prenatal opioid exposure affected GluN2B subunit expression and NMDAR function in rat and chicken cerebellum. Pregnant rats were exposed to methadone (10 mg/kg/day) or buprenorphine (1 mg/kg/day) for the whole period of gestation, using an osmotic minipump. To further examine potential effects of prenatal opioid exposure in a limited time window, chicken embryos were exposed to a 20 mg/kg dose of methadone or morphine on embryonic days 13 and 14. Western blot analysis of cerebella isolated from 14 days old rat pups exposed to buprenorphine showed significantly lower level of the GluN2B subunit, while the opioid exposed chicken embryo cerebellar GluN2B expression remained unaffected at embryonic day 17. However, we observed increased NMDA/glycine-induced calcium influx in cerebellar granule neurone cultures from opioid exposed chicken embryos. We conclude that prenatal opioid exposure leads to opioid receptor-dependent reduction in the postnatal expression of GluN2B in rat cerebella, and increase in NMDA-induced calcium influx in chicken embryo cerebella.
Collapse
Affiliation(s)
- Marthe Fredheim Fjelldal
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Mussie Ghezu Hadera
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Mette Kongstorp
- Section for Drug Abuse Research, Department of Forensic Sciences, Oslo University Hospital, Norway
| | - Lars Peter Engeset Austdal
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Ana Šulović
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Jannike Mørch Andersen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway.,Section for Drug Abuse Research, Department of Forensic Sciences, Oslo University Hospital, Norway
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| |
Collapse
|
35
|
Antonov SA, Novosadova EV, Kobylyansky AG, Illarioshkin SN, Tarantul VZ, Grivennikov IA. Expression and Functional Properties of NMDA and GABA A Receptors during Differentiation of Human Induced Pluripotent Stem Cells into Ventral Mesencephalic Neurons. BIOCHEMISTRY (MOSCOW) 2019; 84:310-320. [PMID: 31221069 DOI: 10.1134/s0006297919030131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ionotropic glutamate and GABA receptors regulate the differentiation and determine the functional properties of mature neurons. Both insufficient and excessive activity of these neurotransmission systems are associated with various nervous system diseases. Our knowledge regarding the expression profiles of these receptors and the mechanisms of their regulation during the differentiation of specialized human neuron subtypes is limited. Here the expression profiles of the NMDA and GABAA receptor subunits were explored during in vitro differentiation of human induced pluripotent stem cells (iPSCs) into ventral mesencephalic neurons. The correlation between the neuronal maturation and the expression dynamics of these genes was investigated, and the functional activity of these receptors was assessed by calcium imaging. The role of NMDA and GABAA receptors in neurite outgrowth and the development of spontaneous activity was analyzed using the viral transduction of neural progenitors with the reporter genes TagGFP and TagRFP. The data indicate that agonists of the investigated receptors can be employed for optimization of existing protocols for neural differentiation of iPSCs, in particular for acceleration of neuronal maturation.
Collapse
Affiliation(s)
- S A Antonov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | - E V Novosadova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - A G Kobylyansky
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | | | - V Z Tarantul
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - I A Grivennikov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| |
Collapse
|
36
|
Szychowski KA, Rombel-Bryzek A, Dołhańczuk-Śródka A, Gmiński J. Antiproliferative Effect of Elastin-Derived Peptide VGVAPG on SH-SY5Y Neuroblastoma Cells. Neurotox Res 2019; 36:503-514. [PMID: 31161598 PMCID: PMC6745029 DOI: 10.1007/s12640-019-00040-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Throughout the lifetime of humans, the amount of stem cells and the rate of cell proliferation continue to decrease. Reactive oxygen species (ROS) are one among the many factors that promote stem cell aging. Both a decrease in the level of stem cells and increase in ROS production can lead to the development of different neurodegenerative diseases. This study was conducted to determine how the VGVAPG peptide, liberated from elastin during the aging process and under pathological conditions, affects ROS production and activities of antioxidant enzymes in undifferentiated, proliferating SH-SY5Y cells. SH-SY5Y cells were maintained in Dulbecco's modified Eagle's medium/nutrient mixture F-12 supplemented with 10% heat-inactivated fetal bovine serum (FBS). After treating the SH-SY5Y cells with VGVAPG peptide, we measured ROS production; cell metabolism, proliferation, and expression; and activities of superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT). We demonstrated that the VGVAPG peptide increases GPx expression and activity, whereas it decreases CAT expression in SH-SY5Y cells. Silencing of the GLB1 gene prevents changes in GPx activity. Despite the fact that the VGVAPG peptide increases GPx expression, it increases the ROS level. Moreover, the VGVAPG peptide decreases SH-SY5Y proliferation, which is prevented by the ROS scavenger N-acetyl-L-cysteine. Our data suggest that ROS production and decreased proliferation of SH-SY5Y cells are the results of excitotoxicity meditated through close unrecognized molecular pathways. More research is needed to elucidate the unknown mechanism of action of VGVAPG peptide in the nervous system.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland.
| | - Agnieszka Rombel-Bryzek
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland
| | | | - Jan Gmiński
- Department of Public Health, Dietetics and Lifestyle Disorders, Faculty of Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszow, Poland
| |
Collapse
|
37
|
SHANK2 mutations associated with autism spectrum disorder cause hyperconnectivity of human neurons. Nat Neurosci 2019; 22:556-564. [PMID: 30911184 PMCID: PMC6475597 DOI: 10.1038/s41593-019-0365-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Heterozygous loss-of-function mutations in SHANK2 are associated with autism spectrum disorder (ASD). We generated cortical neurons from induced pluripotent stem cells (iPSC) derived from neurotypic and ASD-affected donors. We developed Sparse coculture for Connectivity (SparCon) assays where SHANK2 and control neurons were differentially labeled and sparsely seeded together on a lawn of unlabeled control neurons. We observed increases in dendrite length, dendrite complexity, synapse number, and frequency of spontaneous excitatory postsynaptic currents. These findings were phenocopied in gene-edited homozygous SHANK2 knockout cells and rescued by gene correction of an ASD SHANK2 mutation. Dendrite length increases were exacerbated by IGF1, TG003, or BDNF, and suppressed by DHPG treatment. The transcriptome in isogenic SHANK2 neurons was perturbed in synapse, plasticity, and neuronal morphogenesis gene sets and ASD gene modules, and activity-dependent dendrite extension was impaired. Our findings provide evidence for hyperconnectivity and altered transcriptome in SHANK2 neurons derived from ASD subjects.
Collapse
|
38
|
Khalifa D, Shahin O, Salem D, Raafat O. Serum glutamate was elevated in children aged 3-10 years with autism spectrum disorders when they were compared with controls. Acta Paediatr 2019; 108:295-299. [PMID: 29949195 DOI: 10.1111/apa.14477] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 12/03/2017] [Accepted: 06/25/2018] [Indexed: 02/03/2023]
Abstract
AIM This study aimed to see whether measuring serum glutamate in children with autism spectrum disorder (ASD) could provide a biological marker that could allow early intervention. METHODS Serum glutamate was measured in 30 patients aged 3-10 years presenting with ASD to the Abou El Reesh Hospitals, Cairo University, Egypt and 30 matched controls without ASD in 2015. The Vineland Social Maturity Scale was applied to assess social competence, self- help skills and adaptive behaviour in both groups. The severity of autism was measured with the Childhood Autism Rating Scale test. RESULTS The patients' group showed higher mean values of serum glutamate (5.888) than the control group (2.521) and the statistical difference was significant (p = 0.00021). There was no significant difference (p = 0.151) in the serum level of glutamate between patients receiving 1-2 mg of risperidone (6.519 ± 2.851) and those who were free from any medication for at least six weeks (5.157 ± 2.184). CONCLUSION We found higher levels of serum glutamate in subjects with ASD and this might reflect altered glutamatergic neurotransmission which may aid early ASD detection. Further investigations are needed with a large number of participants to further clarify the possibility of using glutamate as a biomarker for ASD.
Collapse
Affiliation(s)
- D Khalifa
- Department of Psychiatry; Faculty of Medicine; Cairo University; Cairo Egypt
| | - O Shahin
- Department of Psychiatry; Faculty of Medicine; Cairo University; Cairo Egypt
| | - D Salem
- Department of Biochemistry; Faculty of Medicine; Cairo University; New Giza University; Cairo Egypt
| | - O Raafat
- Department of Psychiatry; Faculty of Medicine; Cairo University; Cairo Egypt
| |
Collapse
|
39
|
Jeanneret V, Ospina JP, Diaz A, Manrique LG, Merino P, Gutierrez L, Torre E, Wu F, Cheng L, Yepes M. Tissue-type plasminogen activator protects the postsynaptic density in the ischemic brain. J Cereb Blood Flow Metab 2018; 38:1896-1910. [PMID: 29547062 PMCID: PMC6259311 DOI: 10.1177/0271678x18764495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cerebral ischemia causes the presynaptic release of tissue-type plasminogen activator (tPA). The postsynaptic density (PSD) is a postsynaptic structure that provides a matrix where signaling transduction of excitatory synapses takes place. The postsynaptic density protein-95 (PSD-95) is the most abundant scaffolding protein in the postsynaptic density (PSD), where it modulates the postsynaptic response to the presynaptic release of glutamate by regulating the anchoring of glutamate receptors to the PSD. We found that tPA induces the local translation of PSD-95 mRNA and the subsequent recruitment of PSD-95 protein to the PSD, via plasminogen-independent activation of TrkB receptors. Our data show that PSD-95 is removed from the PSD during the early stages of cerebral ischemia, and that this effect is abrogated by either the release of neuronal tPA, or intravenous administration of recombinant tPA (rtPA). We report that the effect of tPA on PSD-95 is associated with inhibition of the phosphorylation and recruitment of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors to the PSD, known to amplify the effect of the excitotoxic injury, and that this is followed by TrkB-mediated protection of dendritic spines from the harmful effects of the hypoxic insult. These data reveal that tPA is a synaptic protector in the ischemic brain.
Collapse
Affiliation(s)
- Valerie Jeanneret
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Juan P Ospina
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ariel Diaz
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Luis G Manrique
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Paola Merino
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Laura Gutierrez
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Enrique Torre
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Fang Wu
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Lihong Cheng
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Manuel Yepes
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA.,3 Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| |
Collapse
|
40
|
Jakaria M, Park SY, Haque ME, Karthivashan G, Kim IS, Ganesan P, Choi DK. Neurotoxic Agent-Induced Injury in Neurodegenerative Disease Model: Focus on Involvement of Glutamate Receptors. Front Mol Neurosci 2018; 11:307. [PMID: 30210294 PMCID: PMC6123546 DOI: 10.3389/fnmol.2018.00307] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022] Open
Abstract
Glutamate receptors play a crucial role in the central nervous system and are implicated in different brain disorders. They play a significant role in the pathogenesis of neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Although many studies on NDDs have been conducted, their exact pathophysiological characteristics are still not fully understood. In in vivo and in vitro models of neurotoxic-induced NDDs, neurotoxic agents are used to induce several neuronal injuries for the purpose of correlating them with the pathological characteristics of NDDs. Moreover, therapeutic drugs might be discovered based on the studies employing these models. In NDD models, different neurotoxic agents, namely, kainic acid, domoic acid, glutamate, β-N-Methylamino-L-alanine, amyloid beta, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, 1-methyl-4-phenylpyridinium, rotenone, 3-Nitropropionic acid and methamphetamine can potently impair both ionotropic and metabotropic glutamate receptors, leading to the progression of toxicity. Many other neurotoxic agents mainly affect the functions of ionotropic glutamate receptors. We discuss particular neurotoxic agents that can act upon glutamate receptors so as to effectively mimic NDDs. The correlation of neurotoxic agent-induced disease characteristics with glutamate receptors would aid the discovery and development of therapeutic drugs for NDDs.
Collapse
Affiliation(s)
- Md. Jakaria
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Shin-Young Park
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Md. Ezazul Haque
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | - Govindarajan Karthivashan
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - In-Su Kim
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - Palanivel Ganesan
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
- Nanotechnology Research Center, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
- Nanotechnology Research Center, Konkuk University, Chungju, South Korea
| |
Collapse
|
41
|
Boente-Juncal A, Méndez AG, Vale C, Vieytes MR, Botana LM. In Vitro Effects of Chronic Spirolide Treatment on Human Neuronal Stem Cell Differentiation and Cholinergic System Development. ACS Chem Neurosci 2018. [PMID: 29518322 DOI: 10.1021/acschemneuro.8b00036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spirolides (SPX) are marine toxins, produced by dinoflagellates that act as potent antagonists of nicotinic acetylcholine receptors. These compounds are not toxic for humans, and since there are no reports of human intoxications caused by this group of toxins they are not yet currently regulated in Europe. Currently 13-desmethyl spirolide C, 13,19-didesmethyl spirolide C, and 20-methyl spirolide G are commercially available as reference materials. Previous work in our laboratory has demonstrated that after 4 days of treatment of primary mice cortical neurons with 13-desmethyl spirolide C, the compound ameliorated the glutamate induced toxicity and increased acetylcholine levels and the expression of the acetylcholine synthesizing enzyme being useful both in vitro and in vivo to decrease the brain pathology associated with Alzheimer's disease. In this work, we aimed to extend the study of the neuronal effects of spirolides in human neuronal cells. To this end, human neuronal progenitor cells CTX0E16 were employed to evaluate the in vitro effect of spirolides on neuronal development. The results presented here indicate that long-term exposure (30 days) of human neuronal stem cells to SPX compounds, at concentrations up to 50 nM, ameliorated the MPP+-induced neurotoxicity and increased the expression of neuritic and dendritic markers, the levels of the choline acetyltransferase enzyme and the protein levels of the α7 subunit of nicotinic acetylcholine receptors. These effects are presumably due to the previously described interaction of these compounds with nicotinic receptors containing both α7 and α4 subunits. All together, these data emphasize the idea that SPX could be attractive lead molecules against neurodegenerative disorders.
Collapse
Affiliation(s)
- Andrea Boente-Juncal
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27001 Lugo, Spain
| | - Aida G. Méndez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27001 Lugo, Spain
| | - Carmen Vale
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27001 Lugo, Spain
| | - Mercedes R. Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27001 Lugo, Spain
| | - Luis M. Botana
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27001 Lugo, Spain
| |
Collapse
|
42
|
Torres-Rojas C, Jones BC. Sex Differences in Neurotoxicogenetics. Front Genet 2018; 9:196. [PMID: 29922331 PMCID: PMC5996082 DOI: 10.3389/fgene.2018.00196] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/15/2018] [Indexed: 12/02/2022] Open
Abstract
A major development in biomedical research is the recognition that the sex of an individual plays a key role in susceptibility, treatment, and outcomes of most diseases. In this contribution, we present evidence that sex is also important in the toxicity of many environmental toxicants and contributes to the effect of genetics. Thus, individual differences in response to toxicants includes genetic makeup, the environment and sex; in fact, sex differences may be considered a part of genetic constitution. In this review, we present evidence for sex contribution to susceptibility for a number of toxicants.
Collapse
Affiliation(s)
- Carolina Torres-Rojas
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
43
|
Jahanbazi Jahan-Abad A, Sahab Negah S, Hosseini Ravandi H, Ghasemi S, Borhani-Haghighi M, Stummer W, Gorji A, Khaleghi Ghadiri M. Human Neural Stem/Progenitor Cells Derived From Epileptic Human Brain in a Self-Assembling Peptide Nanoscaffold Improve Traumatic Brain Injury in Rats. Mol Neurobiol 2018; 55:9122-9138. [PMID: 29651746 DOI: 10.1007/s12035-018-1050-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/27/2018] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is a disruption in the brain functions following a head trauma. Cell therapy may provide a promising treatment for TBI. Among different cell types, human neural stem cells cultured in self-assembling peptide scaffolds have been suggested as a potential novel method for cell replacement treatment after TBI. In the present study, we accessed the effects of human neural stem/progenitor cells (hNS/PCs) derived from epileptic human brain and human adipose-derived stromal/stem cells (hADSCs) seeded in PuraMatrix hydrogel (PM) on brain function after TBI in an animal model of brain injury. hNS/PCs were isolated from patients with medically intractable epilepsy undergone epilepsy surgery. hNS/PCs and hADSCs have the potential for proliferation and differentiation into both neuronal and glial lineages. Assessment of the growth characteristics of hNS/PCs and hADSCs revealed that the hNS/PCs doubling time was significantly longer and the growth rate was lower than hADSCs. Transplantation of hNS/PCs and hADSCs seeded in PM improved functional recovery, decreased lesion volume, inhibited neuroinflammation, and reduced the reactive gliosis at the injury site. The data suggest the transplantation of hNS/PCs or hADSCs cultured in PM as a promising treatment option for cell replacement therapy in TBI.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sedigheh Ghasemi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran. .,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Robert-Koch-Strasse 45, 48149, Münster, Germany.
| | | |
Collapse
|
44
|
Gibson CL, Balbona JT, Niedzwiecki A, Rodriguez P, Nguyen KCQ, Hall DH, Blakely RD. Glial loss of the metallo β-lactamase domain containing protein, SWIP-10, induces age- and glutamate-signaling dependent, dopamine neuron degeneration. PLoS Genet 2018; 14:e1007269. [PMID: 29590100 PMCID: PMC5891035 DOI: 10.1371/journal.pgen.1007269] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/09/2018] [Accepted: 02/22/2018] [Indexed: 12/24/2022] Open
Abstract
Across phylogeny, glutamate (Glu) signaling plays a critical role in regulating neural excitability, thus supporting many complex behaviors. Perturbed synaptic and extrasynaptic Glu homeostasis in the human brain has been implicated in multiple neuropsychiatric and neurodegenerative disorders including Parkinson's disease, where theories suggest that excitotoxic insults may accelerate a naturally occurring process of dopamine (DA) neuron degeneration. In C. elegans, mutation of the glial expressed gene, swip-10, results in Glu-dependent DA neuron hyperexcitation that leads to elevated DA release, triggering DA signaling-dependent motor paralysis. Here, we demonstrate that swip-10 mutations induce premature and progressive DA neuron degeneration, with light and electron microscopy studies demonstrating the presence of dystrophic dendritic processes, as well as shrunken and/or missing cell soma. As with paralysis, DA neuron degeneration in swip-10 mutants is rescued by glial-specific, but not DA neuron-specific expression of wildtype swip-10, consistent with a cell non-autonomous mechanism. Genetic studies implicate the vesicular Glu transporter VGLU-3 and the cystine/Glu exchanger homolog AAT-1 as potential sources of Glu signaling supporting DA neuron degeneration. Degeneration can be significantly suppressed by mutations in the Ca2+ permeable Glu receptors, nmr-2 and glr-1, in genes that support intracellular Ca2+ signaling and Ca2+-dependent proteolysis, as well as genes involved in apoptotic cell death. Our studies suggest that Glu stimulation of nematode DA neurons in early larval stages, without the protective actions of SWIP-10, contributes to insults that ultimately drive DA neuron degeneration. The swip-10 model may provide an efficient platform for the identification of molecular mechanisms that enhance risk for Parkinson's disease and/or the identification of agents that can limit neurodegenerative disease progression.
Collapse
Affiliation(s)
- Chelsea L. Gibson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
| | - Joseph T. Balbona
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
| | - Ashlin Niedzwiecki
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
| | - Peter Rodriguez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
| | - Ken C. Q. Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Randy D. Blakely
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
- Department of Psychiatry, Vanderbilt University, Nashville, TN, United States of America
- The Brain Institute, Florida Atlantic University, Jupiter, FL, United States of America
- * E-mail:
| |
Collapse
|
45
|
Jha SK, Jha NK, Kumar D, Sharma R, Shrivastava A, Ambasta RK, Kumar P. Stress-Induced Synaptic Dysfunction and Neurotransmitter Release in Alzheimer's Disease: Can Neurotransmitters and Neuromodulators be Potential Therapeutic Targets? J Alzheimers Dis 2018; 57:1017-1039. [PMID: 27662312 DOI: 10.3233/jad-160623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The communication between neurons at synaptic junctions is an intriguing process that monitors the transmission of various electro-chemical signals in the central nervous system. Albeit any aberration in the mechanisms associated with transmission of these signals leads to loss of synaptic contacts in both the neocortex and hippocampus thereby causing insidious cognitive decline and memory dysfunction. Compelling evidence suggests that soluble amyloid-β (Aβ) and hyperphosphorylated tau serve as toxins in the dysfunction of synaptic plasticity and aberrant neurotransmitter (NT) release at synapses consequently causing a cognitive decline in Alzheimer's disease (AD). Further, an imbalance between excitatory and inhibitory neurotransmission systems induced by impaired redox signaling and altered mitochondrial integrity is also amenable for such abnormalities. Defective NT release at the synaptic junction causes several detrimental effects associated with altered activity of synaptic proteins, transcription factors, Ca2+ homeostasis, and other molecules critical for neuronal plasticity. These detrimental effects further disrupt the normal homeostasis of neuronal cells and thereby causing synaptic loss. Moreover, the precise mechanistic role played by impaired NTs and neuromodulators (NMs) and altered redox signaling in synaptic dysfunction remains mysterious, and their possible interlink still needs to be investigated. Therefore, this review elucidates the intricate role played by both defective NTs/NMs and altered redox signaling in synaptopathy. Further, the involvement of numerous pharmacological approaches to compensate neurotransmission imbalance has also been discussed, which may be considered as a potential therapeutic approach in synaptopathy associated with AD.
Collapse
|
46
|
Vitamin E-Mediated Modulation of Glutamate Receptor Expression in an Oxidative Stress Model of Neural Cells Derived from Embryonic Stem Cell Cultures. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:6048936. [PMID: 29348770 PMCID: PMC5733950 DOI: 10.1155/2017/6048936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/05/2017] [Accepted: 10/29/2017] [Indexed: 12/23/2022]
Abstract
Glutamate is the primary excitatory neurotransmitter in the central nervous system. Excessive concentrations of glutamate in the brain can be excitotoxic and cause oxidative stress, which is associated with Alzheimer's disease. In the present study, the effects of vitamin E in the form of tocotrienol-rich fraction (TRF) and alpha-tocopherol (α-TCP) in modulating the glutamate receptor and neuron injury markers in an in vitro model of oxidative stress in neural-derived embryonic stem (ES) cell cultures were elucidated. A transgenic mouse ES cell line (46C) was differentiated into a neural lineage in vitro via induction with retinoic acid. These cells were then subjected to oxidative stress with a significantly high concentration of glutamate. Measurement of reactive oxygen species (ROS) was performed after inducing glutamate excitotoxicity, and recovery from this toxicity in response to vitamin E was determined. The gene expression levels of glutamate receptors and neuron-specific enolase were elucidated using real-time PCR. The results reveal that neural cells derived from 46C cells and subjected to oxidative stress exhibit downregulation of NMDA, kainate receptor, and NSE after posttreatment with different concentrations of TRF and α-TCP, a sign of neurorecovery. Treatment of either TRF or α-TCP reduced the levels of ROS in neural cells subjected to glutamate-induced oxidative stress; these results indicated that vitamin E is a potent antioxidant.
Collapse
|
47
|
Pons-Bennaceur A, Lozovaya N. Electrophysiological Investigation of NMDA Current Properties in Brain Slices. Methods Mol Biol 2017; 1677:231-239. [PMID: 28986876 DOI: 10.1007/978-1-4939-7321-7_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Analysis of electrophysiological properties of NMDARs and NMDAR-mediated synaptic transmission in identified neurons and synapses in brain slices is a major step in understanding their function in normal and pathological neuronal brain networks. In many central synapses excitatory postsynaptic currents (EPSCs) are mediated by excitatory neurotransmitter glutamate that activates colocalized AMPAR and NMDAR generating a complex EPSC. Here, we describe the methods commonly used in brain slices to study the electrophysiological properties of NMDAR-mediated component of spontaneous or evoked EPSCs by extracellular stimulation or by stimulating synaptically connected neurons. This approach is based on whole-cell patch-clamp recordings, pharmacological tools, and the analysis of the difference in temporal parameters between the AMPA and NMDA receptors. It allows pinpointing of the basic functional properties of NMDARs that are specific to identified brain regions, neurons, and synapses of wild-type or genetically manipulated mice.
Collapse
Affiliation(s)
- Alexandre Pons-Bennaceur
- INSERM U901, Mediterranean Institute of Neurobiology (INMED), Aix-Marseille University, Marseille, France
| | - Natalia Lozovaya
- Neurochlore c/o Mediterranean Institute of Neurobiology (INMED), INSERM U901, Parc Scientifique de Luminy, BP13, 13273, Marseille Cedex 09, France.
| |
Collapse
|
48
|
Xu JC, Fan J, Wang X, Eacker SM, Kam TI, Chen L, Yin X, Zhu J, Chi Z, Jiang H, Chen R, Dawson TM, Dawson VL. Cultured networks of excitatory projection neurons and inhibitory interneurons for studying human cortical neurotoxicity. Sci Transl Med 2016; 8:333ra48. [PMID: 27053772 DOI: 10.1126/scitranslmed.aad0623] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 03/18/2016] [Indexed: 12/12/2022]
Abstract
Translating neuroprotective treatments from discovery in cell and animal models to the clinic has proven challenging. To reduce the gap between basic studies of neurotoxicity and neuroprotection and clinically relevant therapies, we developed a human cortical neuron culture system from human embryonic stem cells or human inducible pluripotent stem cells that generated both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. This methodology used timed administration of retinoic acid to FOXG1(+) neural precursor cells leading to differentiation of neuronal populations representative of the six cortical layers with both excitatory and inhibitory neuronal networks that were functional and homeostatically stable. In human cortical neuronal cultures, excitotoxicity or ischemia due to oxygen and glucose deprivation led to cell death that was dependent on N-methyl-D-aspartate (NMDA) receptors, nitric oxide (NO), and poly(ADP-ribose) polymerase (PARP) (a cell death pathway called parthanatos that is distinct from apoptosis, necroptosis, and other forms of cell death). Neuronal cell death was attenuated by PARP inhibitors that are currently in clinical trials for cancer treatment. This culture system provides a new platform for the study of human cortical neurotoxicity and suggests that PARP inhibitors may be useful for ameliorating excitotoxic and ischemic cell death in human neurons.
Collapse
Affiliation(s)
- Jin-Chong Xu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jing Fan
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xueqing Wang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen M Eacker
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Li Chen
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiling Yin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juehua Zhu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, Jiangsu 210002, China
| | - Zhikai Chi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Haisong Jiang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rong Chen
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
49
|
Hill E, Nagel D, Parri R, Coleman M. Stem cell-derived astrocytes: are they physiologically credible? J Physiol 2016; 594:6595-6606. [PMID: 26634807 PMCID: PMC5108894 DOI: 10.1113/jp270658] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/28/2015] [Indexed: 01/29/2023] Open
Abstract
Astrocytes are now increasingly acknowledged as having fundamental and sophisticated roles in brain function and dysfunction. Unravelling the complex mechanisms that underlie human brain astrocyte-neuron interactions is therefore an essential step on the way to understanding how the brain operates. Insights into astrocyte function to date have almost exclusively been derived from studies conducted using murine or rodent models. Whilst these have led to significant discoveries, preliminary work with human astrocytes has revealed a hitherto unknown range of astrocyte types with potentially greater functional complexity and increased neuronal interaction with respect to animal astrocytes. It is becoming apparent, therefore, that many important functions of astrocytes will only be discovered by direct physiological interrogation of human astrocytes. Recent advancements in the field of stem cell biology have provided a source of human-based models. These will provide a platform to facilitate our understanding of normal astrocyte functions as well as their role in CNS pathology. A number of recent studies have demonstrated that stem cell-derived astrocytes exhibit a range of properties, suggesting that they may be functionally equivalent to their in vivo counterparts. Further validation against in vivo models will ultimately confirm the future utility of these stem cell-based approaches in fulfilling the need for human-based cellular models for basic and clinical research. In this review we discuss the roles of astrocytes in the brain and highlight the extent to which human stem cell-derived astrocytes have demonstrated functional activities that are equivalent to those observed in vivo.
Collapse
Affiliation(s)
- Eric Hill
- Aston Research Centre for Healthy AgeingLife and Health SciencesAston UniversityBirminghamB4 7ETUK
| | - David Nagel
- Aston Research Centre for Healthy AgeingLife and Health SciencesAston UniversityBirminghamB4 7ETUK
| | - Rheinallt Parri
- Aston Research Centre for Healthy AgeingLife and Health SciencesAston UniversityBirminghamB4 7ETUK
| | - Michael Coleman
- Aston Research Centre for Healthy AgeingLife and Health SciencesAston UniversityBirminghamB4 7ETUK
| |
Collapse
|
50
|
Zhang H, Gelernter J. Review: DNA methylation and alcohol use disorders: Progress and challenges. Am J Addict 2016; 26:502-515. [PMID: 27759945 DOI: 10.1111/ajad.12465] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/17/2016] [Accepted: 10/02/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Risk for alcohol use disorders (AUDs) is influenced by gene-environment interactions. Environmental factors can affect gene expression through epigenetic mechanisms such as DNA methylation. This review outlines the findings regarding the association of DNA methylation and AUDs. METHODS We searched PubMed (by April 2016) and identified 29 studies that examined the association of DNA methylation and AUDs. We also evaluated the methods used in these studies. RESULTS Two studies demonstrated elevated global (repetitive element) DNA methylation levels in AUD subjects. Fifteen candidate gene studies showed hypermethylation of promoter regions of six genes (AVP, DNMT3B, HERP, HTR3A, OPRM1, and SNCA) or hypomethylation of the GDAP1 promoter region in AUD subjects. Five genome-wide DNA methylation studies demonstrated widespread DNA methylation changes across the genome in AUD subjects. Six studies showed significant correlations of DNA methylation with gene expression in AUD subjects. Three studies revealed interactive effects of genetic variation and DNA methylation on susceptibility to AUDs. Most studies analyzed AUD-associated DNA methylation changes in the peripheral blood; a few studies examined DNA methylation changes in postmortem brains of AUD subjects. DISCUSSION AND CONCLUSIONS Chronic alcohol consumption may result in DNA methylation changes, leading to neuroadaptations that may underlie some of the mechanisms of AUD risk and persistence. Future studies are needed to confirm the few existing results, and then to elucidate whether DNA methylation changes are the cause or consequence of AUDs. SCIENTIFIC SIGNIFICANCE DNA methylation profiles may be used to assess AUD status or monitor AUD treatment response. (Am J Addict 2017;26:502-515).
Collapse
Affiliation(s)
- Huiping Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,VA Connecticut Healthcare System, West Haven, Connecticut.,Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.,Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|