1
|
Zhang X, Cai W, Wang C, Tian J. N-methyl-d-aspartate receptors (NMDARs): a glutamate-activated cation channel with biased signaling and therapeutic potential in brain disorders. Pharmacol Ther 2025:108888. [PMID: 40412765 DOI: 10.1016/j.pharmthera.2025.108888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/21/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are a type of calcium-permeable ion channel receptors that are extensively distributed throughout the body, composed of various subunits. The presence of diverse ligands and subcellular localizations of the receptors confer biased signaling and distinct functional roles. Activation of the NMDARs induces calcium influx, which plays a pivotal role in neurotransmitter release, synaptic plasticity, and intracellular signaling. Differential localization of NMDARs at synaptic and extrasynaptic sites results in divergent physiological effects; excessive or insufficient activation of NMDARs disrupts calcium homeostasis, leading to neuronal damage and subsequent neurological dysfunction as well as related diseases. Therefore, it is crucial to develop drugs targeting NMDAR with high efficacy with low toxicity for treating disorders associated with NMDARs abnormalities. In this review, we summarize both fundamental and clinical studies on NMDARs while discussing potential therapeutic targets aimed at modulating ion channel activity through regulating mechanisms, subunit rearrangement, membrane expression, and the specific targeting of synaptic versus extrasynaptic NMDARs. Our goal is to provide new insights for innovative drug development.
Collapse
Affiliation(s)
- Xuan Zhang
- Institute of Brain Disease and Data Analysis, College of Life Sciences and Oceanography, Shenzhen University, 518060, Guangdong, PR China
| | - Wensheng Cai
- Institute of Brain Disease and Data Analysis, College of Life Sciences and Oceanography, Shenzhen University, 518060, Guangdong, PR China
| | - Chao Wang
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, Guang Dong, PR China
| | - Jing Tian
- Institute of Brain Disease and Data Analysis, College of Life Sciences and Oceanography, Shenzhen University, 518060, Guangdong, PR China.
| |
Collapse
|
2
|
Do Carmo S, Kautzmann MAI, Bhattacharjee S, Jun B, Steinberg C, Emmerson JT, Malcolm JC, Bonomo Q, Bazan NG, Cuello AC. Differential effect of an evolving amyloid and tau pathology on brain phospholipids and bioactive lipid mediators in rat models of Alzheimer-like pathology. J Neuroinflammation 2024; 21:185. [PMID: 39080670 PMCID: PMC11290283 DOI: 10.1186/s12974-024-03184-7] [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: 04/19/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Brain inflammation contributes significantly to the pathophysiology of Alzheimer's disease, and it is manifested by glial cell activation, increased production of cytokines/chemokines, and a shift in lipid mediators from a pro-homeostatic to a pro-inflammatory profile. However, whether the production of bioactive lipid mediators is affected at earlier stages, prior to the deposition of Aβ plaques and tau hyperphosphorylation, is unknown. The differential contribution of an evolving amyloid and tau pathology on the composition and abundance of membrane phospholipids and bioactive lipid mediators also remains unresolved. METHODS In this study, we examined the cortical levels of DHA- and AA-derived bioactive lipid mediators and of membrane phospholipids by liquid chromatography with tandem mass spectrometry in transgenic rat models of the Alzheimer's-like amyloid and tau pathologies at early and advanced pathological stages. RESULTS Our findings revealed a complex balance between pro-inflammatory and pro-resolving processes in which tau pathology has a more pronounced effect compared to amyloid pathology. At stages preceding tau misfolding and aggregation, there was an increase in pro-resolving lipid mediators (RVD6 and NPD1), DHA-containing phospholipids and IFN-γ levels. However, in advanced tau pathology displaying NFT-like inclusions, neuronal death, glial activation and cognitive deficits, there was an increase in cytokine and PGD2, PGE2, and PGF2α generation accompanied by a drop in IFN-γ levels. This pathology also resulted in a marked increase in AA-containing phospholipids. In comparison, pre-plaque amyloid pathology already presented high levels of cytokines and AA-containing phospholipids together with elevated RVD6 and NPD1 levels. Finally, Aβ plaque deposition was accompanied by a modest increase in prostaglandins, increased AA-containing phospholipids and reduced DHA-containing phospholipids. CONCLUSIONS Our findings suggest a dynamic trajectory of inflammatory and lipid mediators in the evolving amyloid and tau pathologies and support their differing roles on membrane properties and, consequentially, on signal transduction.
Collapse
Affiliation(s)
- Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
| | - Marie-Audrey I Kautzmann
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Carolyn Steinberg
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Joshua T Emmerson
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada
| | - Janice C Malcolm
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada
| | - Quentin Bonomo
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada
| | - Nicolas G Bazan
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA.
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1210, Montreal, H3G 1Y6, Canada.
- Department of Cell Anatomy and Cell Biology, McGill University, Montreal, H3A 0C7, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, H3G 1Y6, Canada.
- Department of Pharmacology, Oxford University, Oxford, OX1 3QT, UK.
| |
Collapse
|
3
|
Stachowicz K, Pańczyszyn-Trzewik P, Misztak P, Rzeźniczek S, Sowa-Kućma M. Cyclooxygenase-2 inhibition affects the ratio of GluN2A/GluN2B receptor subunits through interaction with mGluR5 in the mouse brain. Neuropeptides 2024; 104:102409. [PMID: 38244260 DOI: 10.1016/j.npep.2024.102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
N-methyl-D-aspartic acid receptors (NMDARs) are the most studied receptors in mammalian brains. Their role in depression, cognition, schizophrenia, learning and memorization, Alzheimer's disease, and more is well documented. In the search for new drug candidates in depression, intensive studies have been conducted. Compounds that act by influencing NMDARs have been particularly intensively investigated following the success of ketamine in clinics. Unfortunately, the side effects associated with ketamine do not allow it to be useful in all cases. Therefore, it is important to learn about new unknown mechanisms related to NMDAR activation and study the impact of changes in the excitatory synapse environment on this receptor. Both direct and intermediary influence on NMDARs via mGluRs and COX-2 are effective. Our prior studies showed that both mGluRs ligands and COX-2 inhibitors are potent in depression-like and cognitive studies through mutual interactions. The side effects associated with imipramine administration, e.g., memory impairment, were improved when inhibiting COX-2. Therefore, this study is a trial that involves searching for modifications in NMDARs in mouse brains after prolonged treatment with MTEP (mGluR5 antagonist), NS398 (COX-2 inhibitor), or imipramine (tricyclic antidepressant). The prefrontal cortex (PFC) and hippocampus (HC) were selected for PCR and Western blot analyses. Altered expression of Gin2a or Grin2b genes after treatment was found. The observed effects were more potent when COX-2 was inhibited. The finding described here may be vital when searching for new drugs acting via NMDARs without the side effects related to cognition.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland.
| | - Patrycja Pańczyszyn-Trzewik
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| | - Paulina Misztak
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland
| | - Szymon Rzeźniczek
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland
| | - Magdalena Sowa-Kućma
- Medical College of Rzeszów University, Institute of Medical Sciences, Department of Human Physiology, 35-310 Rzeszow, Kopisto Street 2a, Poland
| |
Collapse
|
4
|
Stachowicz K. Deciphering the mechanisms of reciprocal regulation or interdependence at the cannabinoid CB1 receptors and cyclooxygenase-2 level: Effects on mood, cognitive implications, and synaptic signaling. Neurosci Biobehav Rev 2023; 155:105439. [PMID: 37898448 DOI: 10.1016/j.neubiorev.2023.105439] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
The lipid endocannabinoid system refers to endogenous cannabinoids (eCBs), the enzymes involved in their synthesis and metabolism, and the G protein-coupled cannabinoid receptors (GPCRs), CB1, and CB2. CB1 receptors (CB1Rs) are distributed in the brain at presynaptic terminals. Their activation induces inhibition of neurotransmitter release, which are gamma-aminobutyric acid (GABA), glutamate (Glu), dopamine, norepinephrine, serotonin, and acetylcholine. Postsynaptically localized CB1Rs regulate the activity of selected ion channels and N-methyl-D-aspartate receptors (NMDARs). CB2Rs are mainly peripheral and will not be considered here. Anandamide metabolism, mediated by cyclooxygenase-2 (COX-2), generates anandamide-derived prostanoids. In addition, COX-2 regulates the formation of CB1 ligands, which reduce excitatory transmission in the hippocampus (HC). The role of CB1Rs and COX-2 has been described in anxiety, depression, and cognition, among other central nervous system (CNS) disorders, affecting neurotransmission and behavior of the synapses. This review will analyze common pathways, mechanisms, and behavioral effects of manipulation at the CB1Rs/COX-2 level.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacoslogy, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| |
Collapse
|
5
|
Sharma R, Neupane C, Pham TL, Lee M, Lee S, Lee SY, Nam MH, Kim CS, Park JB. Tonic Activation of NR2D-Containing NMDARs Exacerbates Dopaminergic Neuronal Loss in MPTP-Injected Parkinsonian Mice. J Neurosci 2023; 43:7730-7744. [PMID: 37726169 PMCID: PMC10648527 DOI: 10.1523/jneurosci.1955-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
NR2D subunit-containing NMDA receptors (NMDARs) gradually disappear during brain maturation but can be recruited by pathophysiological stimuli in the adult brain. Here, we report that 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication recruited NR2D subunit-containing NMDARs that generated an Mg2+-resistant tonic NMDA current (INMDA) in dopaminergic (DA) neurons in the midbrain of mature male mice. MPTP selectively generated an Mg2+-resistant tonic INMDA in DA neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). Consistently, MPTP increased NR2D but not NR2B expression in the midbrain regions. Pharmacological or genetic NR2D interventions abolished the generation of Mg2+-resistant tonic INMDA in SNpc DA neurons, and thus attenuated subsequent DA neuronal loss and gait deficits in MPTP-treated mice. These results show that extrasynaptic NR2D recruitment generates Mg2+-resistant tonic INMDA and exacerbates DA neuronal loss, thus contributing to MPTP-induced Parkinsonism. The state-dependent NR2D recruitment could be a novel therapeutic target for mitigating cell type-specific neuronal death in neurodegenerative diseases.SIGNIFICANCE STATEMENT NR2D subunit-containing NMDA receptors (NMDARs) are widely expressed in the brain during late embryonic and early postnatal development, and then downregulated during brain maturation and preserved at low levels in a few regions of the adult brain. Certain stimuli can recruit NR2D subunits to generate tonic persistent NMDAR currents in nondepolarized neurons in the mature brain. Our results show that MPTP intoxication recruits NR2D subunits in midbrain dopaminergic (DA) neurons, which leads to tonic NMDAR current-promoting dopaminergic neuronal death and consequent abnormal gait behavior in the MPTP mouse model of Parkinson's disease (PD). This is the first study to indicate that extrasynaptic NR2D recruitment could be a target for preventing neuronal death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ramesh Sharma
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Chiranjivi Neupane
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Thuy Linh Pham
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Miae Lee
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Cuk-Seong Kim
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| |
Collapse
|
6
|
Nelson ED, Maynard KR, Nicholas KR, Tran MN, Divecha HR, Collado-Torres L, Hicks SC, Martinowich K. Activity-regulated gene expression across cell types of the mouse hippocampus. Hippocampus 2023; 33:1009-1027. [PMID: 37226416 PMCID: PMC11129873 DOI: 10.1002/hipo.23548] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/19/2023] [Accepted: 05/06/2023] [Indexed: 05/26/2023]
Abstract
Activity-regulated gene (ARG) expression patterns in the hippocampus (HPC) regulate synaptic plasticity, learning, and memory, and are linked to both risk and treatment responses for many neuropsychiatric disorders. The HPC contains discrete classes of neurons with specialized functions, but cell type-specific activity-regulated transcriptional programs are not well characterized. Here, we used single-nucleus RNA-sequencing (snRNA-seq) in a mouse model of acute electroconvulsive seizures (ECS) to identify cell type-specific molecular signatures associated with induced activity in HPC neurons. We used unsupervised clustering and a priori marker genes to computationally annotate 15,990 high-quality HPC neuronal nuclei from N = 4 mice across all major HPC subregions and neuron types. Activity-induced transcriptomic responses were divergent across neuron populations, with dentate granule cells being particularly responsive to activity. Differential expression analysis identified both upregulated and downregulated cell type-specific gene sets in neurons following ECS. Within these gene sets, we identified enrichment of pathways associated with varying biological processes such as synapse organization, cellular signaling, and transcriptional regulation. Finally, we used matrix factorization to reveal continuous gene expression patterns differentially associated with cell type, ECS, and biological processes. This work provides a rich resource for interrogating activity-regulated transcriptional responses in HPC neurons at single-nuclei resolution in the context of ECS, which can provide biological insight into the roles of defined neuronal subtypes in HPC function.
Collapse
Affiliation(s)
- Erik D. Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kyndall R. Nicholas
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew N Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205
| |
Collapse
|
7
|
Stachowicz K. Regulation of COX-2 expression by selected trace elements and heavy metals: Health implications, and changes in neuronal plasticity. A review. J Trace Elem Med Biol 2023; 79:127226. [PMID: 37257334 DOI: 10.1016/j.jtemb.2023.127226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Trace elements or trace metals are essential components of enzymes, proteins, hormones and play a key role in biochemical processes, cell growth and differentiation, as well as in neurotransmission, affecting human physiology. In nature there are also heavy metals that exhibit toxic effects on the human body, including the brain. The importance of trace elements has been established in neurodegenerative disorders, schizophrenia, depression among others. In parallel, an important regulatory element in the above diseases is cyclooxygenase-2 (COX-2), a modulator of the arachidonic acid (AA) pathway, and a cause of neuroinflammation, and glutamate (Glu) dysregulation, affecting calcium (Ca) metabolism in cells. This review presents the effects of major trace elements and heavy metals on COX-2 expression. Calcium (Ca), zinc (Zn), cadmium (Cd), vanadium (V), nickel (Ni), copper (Cu), and iron (Fe) can potentially increase COX-2 expression, inducing neuroinflammation and Glu excitotoxicity; while magnesium (Mg), lithium (Li), and selenium (Se) can potentially decrease COX-2 expression. The associated mechanisms are described in the article.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| |
Collapse
|
8
|
Schmitt LI, David C, Steffen R, Hezel S, Roos A, Schara-Schmidt U, Kleinschnitz C, Leo M, Hagenacker T. Spinal astrocyte dysfunction drives motor neuron loss in late-onset spinal muscular atrophy. Acta Neuropathol 2023; 145:611-635. [PMID: 36930296 PMCID: PMC10119066 DOI: 10.1007/s00401-023-02554-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023]
Abstract
Spinal muscular atrophy (SMA) is a progressive neuromuscular disorder caused by a loss of the survival of motor neuron 1 (SMN1) gene, resulting in a loss of spinal motor neurons (MNs), leading to muscle weakness and wasting. The pathogenesis of MN loss in SMA and the selective vulnerability in different cellular populations are not fully understood. To investigate the role of spinal astrocytes in the pathogenesis of late-onset SMA, we used a mouse model in addition to in vitro approaches. Immunostaining, Western blot analysis, small interfering ribonucleic acid (siRNA) transfections, functional assays, enzyme-linked immunosorbent assay (ELISA), behavioral tests, and electrophysiological measurements were performed. Early activation of spinal astrocytes and a reduction of the excitatory amino acid transporter 1 (EAAT1) on postnatal day (P) 20 preceded the loss of spinal MNs in SMA mice occurring on P42. EAAT1 reduction resulted in elevated glutamate levels in the spinal cord of SMA mice at P20 and P42. SMA-like astrocytes generated by siRNA and an ex vivo model of glutamate excitotoxicity involving organotypic spinal cord slice cultures revealed the critical role of glutamate homeostasis in the degeneration of MNs. The pre-emptive administration of arundic acid (AA), as an inhibitor of astrocyte activation, to SMA mice prior to the loss of motor neurons (P28) resulted in elevated EAAT1 protein levels compared to vehicle-treated SMA mice and prevented the increase of glutamate in the spinal cord and the loss of spinal MNs. Furthermore, AA preserved motor functions during behavioral experiments, the electrophysiological properties, and muscle alteration of SMA mice. In a translational approach, we transfected healthy human fibroblasts with SMN1 siRNA, resulting in reduced EAAT1 expression and reduced uptake but increased glutamate release. These findings were verified by detecting elevated glutamate levels and reduced levels of EAAT1 in cerebrospinal fluid of untreated SMA type 2 and 3 patients. In addition, glutamate was elevated in serum samples, while EAAT1 was not detectable. Our data give evidence for the crucial role of spinal astrocytes in the pathogenesis of late-onset SMA, a potential driving force for MN loss by glutamate excitotoxicity caused by EAAT1 reduction as an early pathophysiological event. Furthermore, our study introduces EAAT1 as a potential therapeutic target for additional SMN-independent therapy strategies to complement SMN-enhancing drugs.
Collapse
Affiliation(s)
- Linda-Isabell Schmitt
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Christina David
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Rebecca Steffen
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Stefanie Hezel
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Andreas Roos
- Department of Pediatrics 1, Division of Neuropediatrics, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatrics 1, Division of Neuropediatrics, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Markus Leo
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Tim Hagenacker
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| |
Collapse
|
9
|
Chen Y, Holland KD, Shertzer HG, Nebert DW, Dalton TP. Fatal Epileptic Seizures in Mice Having Compromised Glutathione and Ascorbic Acid Biosynthesis. Antioxidants (Basel) 2023; 12:antiox12020448. [PMID: 36830006 PMCID: PMC9952205 DOI: 10.3390/antiox12020448] [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: 01/09/2023] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023] Open
Abstract
Reduced glutathione (GSH) and ascorbic acid (AA) are the two most abundant low-molecular-weight antioxidants in mammalian tissues. GclmKO knockout mice lack the gene encoding the modifier subunit of the rate-limiting enzyme in GSH biosynthesis; GclmKO mice exhibit 10-40% of normal tissue GSH levels and show no overt phenotype. GuloKO knockout mice, lacking a functional Gulo gene encoding L-gulono-γ-lactone oxidase, cannot synthesize AA and depend on dietary ascorbic acid for survival. To elucidate functional crosstalk between GSH and AA in vivo, we generated the GclmKO/GuloKO double-knockout (DKO) mouse. DKO mice exhibited spontaneous epileptic seizures, proceeding to death between postnatal day (PND)14 and PND23. Histologically, DKO mice displayed neuronal loss and glial proliferation in the neocortex and hippocampus. Epileptic seizures and brain pathology in young DKO mice could be prevented with AA supplementation in drinking water (1 g/L). Remarkably, in AA-rescued adult DKO mice, the removal of AA supplementation for 2-3 weeks resulted in similar, but more severe, neocortex and hippocampal pathology and seizures, with death occurring between 12 and 21 days later. These results provide direct evidence for an indispensable, yet underappreciated, role for the interplay between GSH and AA in normal brain function and neuronal health. We speculate that the functional crosstalk between GSH and AA plays an important role in regulating glutamatergic neurotransmission and in protecting against excitotoxicity-induced brain damage.
Collapse
Affiliation(s)
- Ying Chen
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520, USA
- Correspondence: ; Tel.: +1-203-785-4694; Fax: +1-203-724-6023
| | - Katherine D. Holland
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Howard G. Shertzer
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniel W. Nebert
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Departments of Pediatrics and Molecular & Developmental Biology, Cincinnati Children’s Research Center, Cincinnati, OH 45229, USA
| | - Timothy P. Dalton
- Department of Environmental and Public Health Sciences, Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
10
|
Uczay M, Pflüger P, Picada JN, de Oliveira JDM, da SilvaTorres IL, Medeiros HR, Vendruscolo MH, von Poser G, Pereira P. Geniposide and asperuloside alter the COX-2 and GluN2B receptor expression after pilocarpine-induced seizures in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:951-962. [PMID: 36536207 DOI: 10.1007/s00210-022-02367-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022]
Abstract
Asperuloside (ASP) and geniposide (GP) are iridoids that have shown various biological properties, such as reduction of inflammation, oxidative stress, and neuroprotection. The aim of this study was to investigate the mechanism of action of ASP and GP through the experimental model of pilocarpine-induced seizures. Mice were treated daily with saline, valproic acid (VPA), GP (5, 25, or 50 mg/kg), or ASP (20 or 40 mg/kg) for 8 days. Pilocarpine (PILO) treatment was administered after the last day of treatment, and the epileptic behavior was recorded for 1 h and analyzed by an adapted scale. Afterward, the hippocampus and blood samples were collected for western blot analyses, ELISA and comet assay, and bone marrow to the micronucleus test. We evaluated the expression of the inflammatory marker cyclooxygenase-2 (COX-2), GluN2B, a subunit of the NMDA receptor, pGluR1, an AMPA receptor, and the enzyme GAD-1 by western blot and the cytokine TNF-α by ELISA. The treatments with GP and ASP were capable to decrease the latency to the first seizure, although they did not change the latency to status epilepticus (SE). ASP demonstrated a genotoxic potential analyzed by comet assay; however, the micronuclei frequency was not increased in the bone marrow. The GP and ASP treatments were capable to reduce COX-2 and GluN2B receptor expression after PILO exposure. This study suggests that GP and ASP have a protective effect on PILO-induced seizures, decreasing GluN2B receptor and COX-2 expression.
Collapse
Affiliation(s)
- Mariana Uczay
- Laboratory of Neuropharmacology and Preclinical Toxicology, Institute of Basic Health Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil
| | - Pricila Pflüger
- Laboratory of Neuropharmacology and Preclinical Toxicology, Institute of Basic Health Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil
| | | | | | - Iraci Lucena da SilvaTorres
- Laboratory of Pain Pharmacology and Neuromodulation, Federal University of Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil
| | - Helouise Richardt Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation, Federal University of Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil
| | - Maria Helena Vendruscolo
- Laboratory of Pharmacognosy, College of Pharmacy, Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, 90050-170, Brazil
| | - Gilsane von Poser
- Laboratory of Pharmacognosy, College of Pharmacy, Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, 90050-170, Brazil
| | - Patrícia Pereira
- Laboratory of Neuropharmacology and Preclinical Toxicology, Institute of Basic Health Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
11
|
Treviño S, Díaz A, González-López G, Guevara J. Differential biochemical-inflammatory patterns in the astrocyte-neuron axis of the hippocampus and frontal cortex in Wistar rats with metabolic syndrome induced by high fat or carbohydrate diets. J Chem Neuroanat 2022; 126:102186. [PMID: 36374648 DOI: 10.1016/j.jchemneu.2022.102186] [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: 06/18/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
Metabolic syndrome (MetS) is a public health problem and a risk of developing cardiometabolic and neurodegenerative diseases. The biochemical-inflammatory impairment in brain areas related to learning and memory has not been differentiated between MetS models. We aimed to compare the effect of the MetS generated by consuming high-fat (HFD) or -carbohydrate diets (HCD) on the hippocampus and frontal cortex, related to astrocyte-neuron metabolism and neuroinflammation origin. Sixty male Wistar rats were separated into three groups: 1) control group, 2) HCD group, and 3) HFD group. After 3 months, we evaluated zoometry, a serum bioclinical profile, and in the hippocampus and frontal cortex, we performed biochemical assays (concentration of lactate, glutamate, fatty acids, and ASAT, ALAT, and LDH activity), immunoreactivity tests (GFAP, COX2, CD36, and BDNF), and immunoassays (TNF-α, IL-1β, IL-6, and PGE2). The bioclinical parameters showed that both diets induce MetS. At the brain level, it is noteworthy that the HCD group had an increase in lactate and glutamate concentration, reactive astrogliosis, immunoreactive COX2 neurons in the CA1 subfield hippocampus and frontal cortex, and high levels of PGE2, TNF-α, IL-1β, and IL-6, and low BDNF immunoreactivity. Meanwhile, the HFD is highlighted by increased fatty acid levels and CD36 expression in the hippocampus and frontal cortex, strong reactive astrogliosis and COX2 immunoreactivity, and the greatest inflammation with the lowest BDNF immunoreactivity. In conclusion, MetS induction by an HFD or HCD generates different biochemical, cellular, and inflammatory patterns in the hippocampus and frontal cortex.
Collapse
Affiliation(s)
- Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, 14 South. FCQ1, University City, Puebla, C.P. 72560, Mexico.
| | - Alfonso Díaz
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, 22 South. FCQ9, University City, Puebla, C.P. 72560, Mexico
| | - Getsemaní González-López
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, 14 South. FCQ1, University City, Puebla, C.P. 72560, Mexico
| | - Jorge Guevara
- Department of Biochemistry, Faculty of Medicine, University National Autonomous of Mexico, Ciudad de Mexico City, C.P. 04510, Mexico
| |
Collapse
|
12
|
Zhou J, Geng Y, Su T, Wang Q, Ren Y, Zhao J, Fu C, Weber M, Lin H, Kaminker JS, Liu N, Sheng M, Chen Y. NMDA receptor-dependent prostaglandin-endoperoxide synthase 2 induction in neurons promotes glial proliferation during brain development and injury. Cell Rep 2022; 38:110557. [PMID: 35354047 DOI: 10.1016/j.celrep.2022.110557] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/16/2021] [Accepted: 03/01/2022] [Indexed: 12/25/2022] Open
Abstract
Astrocytes play critical roles in brain development and disease, but the mechanisms that regulate astrocyte proliferation are poorly understood. We report that astrocyte proliferation is bi-directionally regulated by neuronal activity via NMDA receptor (NMDAR) signaling in neurons. Prolonged treatment with an NMDAR antagonist reduced expression of cell-cycle-related genes in astrocytes in hippocampal cultures and suppressed astrocyte proliferation in vitro and in vivo, whereas neuronal activation promoted astrocyte proliferation, dependent on neuronal NMDARs. Expression of prostaglandin-endoperoxide synthase 2 (Ptgs2) is induced specifically in neurons by NMDAR activation and is required for activity-dependent astrocyte proliferation through its product, prostaglandin E2 (PGE2). NMDAR inhibition or Ptgs2 genetic ablation in mice reduced the proliferation of astrocytes and microglia induced by mild traumatic brain injury in the absence of secondary excitotoxicity-induced neuronal death. Our study defines an NMDAR-mediated signaling mechanism that allows trans-cellular control of glial proliferation by neurons in brain development and injury.
Collapse
Affiliation(s)
- Jia Zhou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Geng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China
| | - Tonghui Su
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiuyan Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China
| | - Yongfei Ren
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zhao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaoying Fu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China
| | - Martin Weber
- Department of Neuroscience, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Han Lin
- Department of Neuroscience, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joshua S Kaminker
- Department of Bioinformatics and Computational Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nan Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China
| | - Morgan Sheng
- Department of Neuroscience, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Haike Road, Pudong, Shanghai 201210, China.
| |
Collapse
|
13
|
Application potential of modulation of cyclooxygenase-2 activity: a cognitive approach. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Cognitive functions of the brain depend largely on the condition of the cell membranes and the proportion of fatty acids. It is known and accepted that arachidonic acid (AA) is one of the main ω-6 fatty acids (phospholipids) in brain cells. Metabolism of that fatty acid depends on the functionality and presence of cyclooxygenase (COX). COX is a primary enzyme in the cycle of transformation of AA to prostanoids, which may mediate response of immune cells, contributing to brain function and cognition. Two COX isoforms (COX-1 and COX-2), as well as a splice variant (COX-3), have been detected in the brain. Findings released in the last decade showed that COX-2 may play an important role in cognition. There are many preclinical and clinical reports showing its engagement in Alzheimer disease, spatial learning, and plasticity. This manuscript focuses on summarizing the above-mentioned discoveries.
Collapse
|
14
|
Research Progress on the Role of Inflammatory Mechanisms in the Development of Postoperative Cognitive Dysfunction. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3883204. [PMID: 34869762 PMCID: PMC8642009 DOI: 10.1155/2021/3883204] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/19/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Postoperative cognitive dysfunction (POCD), as one of the common postoperative complications, mainly occurs after surgery and anesthesia, especially in the elderly. It refers to cognitive function changes such as decreased learning and memory ability and inability to concentrate. In severe cases, there could be personality changes and a decline in social behavior. At present, a great deal of research had been carried out on POCD, but its specific mechanism remains unclear. The release of peripheral inflammation-related factors, the degradation and destruction of the blood-brain barrier, the occurrence of central inflammation, and the neuronal apoptosis and synaptic loss could be promoted by neuroinflammation indicating that inflammatory mechanisms may play key roles in the occurrence of POCD.
Collapse
|
15
|
Engin AB, Neagu M. Editorial overview: Neuroreceptors and neurotoxic effect through altered synaptic transmission of neurotransmitters. CURRENT OPINION IN TOXICOLOGY 2021. [DOI: 10.1016/j.cotox.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Upregulation of the mGlu5 receptor and COX-2 protein in the mouse brain after imipramine and NS398, searching for mechanisms of regulation. Neurochem Int 2021; 150:105193. [PMID: 34571049 DOI: 10.1016/j.neuint.2021.105193] [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: 05/13/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/24/2022]
Abstract
Imipramine belongs to a group of tricyclic antidepressants (TCAs). It has been also documented that its antidepressant activity connects with the modulation of cytosolic phospholipase A2 (cPLA2) and arachidonic acid (AA) turnover. Through this mechanism, imipramine can indirectly modify glutamate (Glu) transmission. Additionally, it has been shown that chronic treatment with imipramine results in the upregulation of the metabotropic glutamate receptor subtype 5 (mGlu5 receptor) in the hippocampus of rats. Our previous study revealed that manipulation of the AA pathway via inhibition of cyclooxygenase-2 (COX-2) by selective COX-2 inhibitor (NS398) could effectively modulate the behavior of mice treated with imipramine. Here, we hypothesized that COX-2 inhibition could similarly to imipramine influence mGlu5 receptor, and thus NS398 can modulate the effect of imipramine on Glu. Moreover, such regulation changes should correspond with alterations in neurotransmission. Increased cPLA activity after imipramine administration may change the activity of the AA pathway and the endocannabinoid metabolism, e.g., 2-Arachidonyl-glycerol (2-AG). To verify the idea, mGlu5 receptor level was investigated in the hippocampus (HC) and prefrontal cortex (PFC) of mice treated for 7 or 14 days with imipramine and/or COX-2 inhibitor: NS398. Western blot and PCR analyses were conducted. Moreover, the excitatory (Glu) and inhibitory (gamma-aminobutyric acid; GABA) neurotransmitters were measured using HPLC and 2-AG using ELISA. A time-dependent change in mGlu5 receptor and COX-2 protein level, COX-2 expression, and 2-AG level in the PFC after imipramine administration was found. Up-regulation of mGlu5 receptor after NS398 was found in HC and PFC. A structure-dependent shift between excitatory vs. inhibitory transmission was detected when NS398 and imipramine were co-administered.
Collapse
|
17
|
Deciphering the mechanisms of regulation of an excitatory synapse via cyclooxygenase-2. A review. Biochem Pharmacol 2021; 192:114729. [PMID: 34400127 DOI: 10.1016/j.bcp.2021.114729] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022]
Abstract
Cyclooxygenase (COX) is a heme-containing enzyme that produces prostaglandins (PGs) via a pathway known as the arachidonic acid (AA) cascade. Two isoforms of COX enzyme (COX-1 and COX-2) and splice variant (COX-3) have been described so far. COX-2 is a neuronal enzyme that is intensively produced during activation of the synapse and glutamate (Glu) release. The end product of COX-2 action, prostaglandin E2 (PGE2), regulates Glu level in a retrograde manner. At the same time, the level of Glu, the primary excitatory neurotransmitter, is regulated in the excitatory synapse via Glu receptors, both ionotropic and metabotropic ones. Glu receptors are known modulators of behavior, engaged in cognition and mood. So far, the interaction between ionotropic N-methyl-D-aspartate (NMDA) receptors or metabotropic glutamate (mGluRs) receptors and COX-2 was found. Here, based on literature data and own research, a new mechanism of action of COX-2 in an excitatory synapse will be presented.
Collapse
|
18
|
Mannan A, Garg N, Singh TG, Kang HK. Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ): Molecular Effects and Its Importance as a Novel Therapeutic Target for Cerebral Ischemic Injury. Neurochem Res 2021; 46:2800-2831. [PMID: 34282491 DOI: 10.1007/s11064-021-03402-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-ɣ) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-ɣ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-ɣ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-ɣ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Harmeet Kaur Kang
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
19
|
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
|
20
|
Stachowicz K. Indomethacin, a nonselective cyclooxygenase inhibitor, does not interact with MTEP in antidepressant-like activity, as opposed to imipramine in CD-1 mice. Eur J Pharmacol 2020; 888:173585. [PMID: 32971092 DOI: 10.1016/j.ejphar.2020.173585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/03/2023]
Abstract
The contribution of metabotropic glutamate receptors (mGlu receptors) in depression is well known and tested worldwide. Our previous study showed the involvement of the cyclooxygenase-2 (COX-2) pathway in behavioral changes mediated by an antagonist of metabotropic glutamate receptor subtype 5 (mGlu5 receptor) 3-[(2-methyl-1,3-tiazol-4-yl)ethynyl]-pyridine (MTEP). Among others, we have found that chronic concomitant administration of a COX-2 inhibitor and sub-effective dose of MTEP accelerates antidepressant-like activity of MTEP. This paper seeks to explore whether the same effect would be observed with the use of a non-selective COX inhibitor 2-[1-(4-chlorobenzoyl)-5-methoxy-2-methylindol-3-yl]acetic acid (indomethacin). To that end, we have employed experimental procedure implemented in the earlier research. MTEP and indomethacin or MTEP + indomethacin were used chronically for 7 or 14 days. Then, the Porsolt test, tail suspension test and locomotor activity test were performed. Imipramine was used as a reference compound, as its action is connected with mGlu5 receptor. We found that, in contrast to COX-2 inhibition, indomethacin - acting both through COX-1 and COX-2 - did not release antidepressant-like potential of MTEP. The opposite effect was shown when imipramine was used.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
21
|
López DE, Ballaz SJ. The Role of Brain Cyclooxygenase-2 (Cox-2) Beyond Neuroinflammation: Neuronal Homeostasis in Memory and Anxiety. Mol Neurobiol 2020; 57:5167-5176. [PMID: 32860157 DOI: 10.1007/s12035-020-02087-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Cyclooxygenases are a group of heme-containing isozymes (namely Cox-1 and Cox-2) that catalyze the conversion of arachidonic acid to largely bioactive prostaglandins (PGs). Cox-1 is the ubiquitous housekeeping enzyme, and the mitogen-inducible Cox-2 is activated to cause inflammation. Interestingly, Cox-2 is constitutively expressed in the brain at the postsynaptic dendrites and excitatory terminals of the cortical and spinal cord neurons. Neuronal Cox-2 is activated in response to synaptic excitation to yield PGE2, the predominant Cox-2 metabolite in the brain, which in turn stimulates the release of glutamate and neuronal firing in a retrograde fashion. Cox-2 is also engaged in the metabolism of new endocannabinoids from 2-arachidonoyl-glycerol to modulate their actions at presynaptic terminals. In addition to these interactions, the induction of neuronal Cox-2 is coupled to the trans-synaptic activation of the dopaminergic mesolimbic system and some serotoninergic receptors, which might contribute to the development of emotional behavior. Although much of the focus regarding the induction of Cox-2 in the brain has been centered on neuroinflammation-related neurodegenerative and psychiatric disorders, some evidence also suggests that Cox-2 release during neuronal signaling may be pivotal for the fine tuning of cortical networks to regulate behavior. This review compiles the evidence supporting the homeostatic role of neuronal Cox-2 in synaptic transmission and plasticity, since neuroinflammation is originally triggered by the induction of glial Cox-2 expression. The goal is to provide perspective on the roles of Cox-2 beyond neuroinflammation, such as those played in memory and anxiety, and whose evidence is still scant.
Collapse
Affiliation(s)
- Diana E López
- Biomedical Sciences Graduate Program, Yachay Tech University, Urcuquí, Ecuador
| | - Santiago J Ballaz
- School of Biological Sciences and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí, Ecuador.
| |
Collapse
|
22
|
Huang L, Peng Z, Lu C, Chen Y, Lv JW, Qin M, Liao DF, Liu XM, Shi Z. Ginsenoside Rg1 alleviates repeated alcohol exposure-induced psychomotor and cognitive deficits. Chin Med 2020; 15:44. [PMID: 32411290 PMCID: PMC7206760 DOI: 10.1186/s13020-020-00325-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/28/2020] [Indexed: 12/25/2022] Open
Abstract
Background Chronic alcohol consumption disrupts psychomotor and cognitive functions, most of which are subserved by the dysfunction of hippocampus. Dysregulated excitatory glutamatergic transmission is implicated in repeated alcohol induced psychomotor and cognitive impairment. Ginsenoside Rg1, one of the main active ingredient of the traditional tonic medicine Panax ginseng C.A. Meyer (Araliaceae), has been used to treat cognitive deficits. Particularly, Rg1 has been demonstrated to improve hippocampus-dependent learning in mice and attenuate glutamate-induced excitotoxicity in vitro. Thus, in the present research, we sought to investigate the therapeutic effects of Ginsenoside Rg1 on repeated alcohol induced psychomotor and cognitive deficits in hippocampal-dependent behavioral tasks and unravel the underpinnings of its neuroprotection. Methods Male ICR (CD-1) mice were consecutively intragastrically treated with 20% (w/v) alcohol for 21 days. Then, behavior tests were conducted to evaluate repeated alcohol induced psychomotor and cognitive deficits. Histopathological changes, and biochemical and molecular alterations were assessed to determine the potential neuroprotective mechanism of Rg1. Results The results suggested that Rg1, at the optimal dose of 6 mg/kg, has the potential to ameliorate repeated alcohol induced cognitive deficits by regulating activities of NR2B containing NMDARs and excitotoxic signaling. Conclusion Our findings further provided a new strategy to treat chronic alcohol exposure induced adverse consequences.
Collapse
Affiliation(s)
- Lu Huang
- 1Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, 410208 Hunan China.,2Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632 China
| | - Zhuang Peng
- 1Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, 410208 Hunan China.,5College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Cong Lu
- 3Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193 China
| | - Ying Chen
- 4Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Jing-Wei Lv
- 3Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193 China
| | - Meng Qin
- 5College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Duan-Fang Liao
- 1Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, 410208 Hunan China
| | - Xin-Min Liu
- 1Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, 410208 Hunan China.,3Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193 China
| | - Zhe Shi
- 1Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, 410208 Hunan China
| |
Collapse
|
23
|
NMDARs in Cell Survival and Death: Implications in Stroke Pathogenesis and Treatment. Trends Mol Med 2020; 26:533-551. [PMID: 32470382 DOI: 10.1016/j.molmed.2020.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/22/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
Stroke is a leading cause of death and disability in developed countries. N-methyl-D-aspartate glutamate receptors (NMDARs) have important roles in stroke pathology and recovery. Depending on their subtypes and locations, these NMDARs may promote either neuronal survival or death. Recently, the functions of previously overlooked NMDAR subtypes during stroke were characterized, and NMDARs expressed at different subcellular locations were found to have synergistic rather than opposing functions. Moreover, the complexity of the neuronal survival and death signaling pathways following NMDAR activation was further elucidated. In this review, we summarize the recent developments in these areas and discuss how delineating the dual roles of NMDARs in stroke has directed the development of novel neuroprotective therapeutics for stroke.
Collapse
|
24
|
NS398, a cyclooxygenase-2 inhibitor, reverses memory performance disrupted by imipramine in C57Bl/6J mice. Brain Res 2020; 1734:146741. [PMID: 32088181 DOI: 10.1016/j.brainres.2020.146741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 11/21/2022]
Abstract
Imipramine has been widely used as an antidepressant in the clinic over the years. Unfortunately, it produces a detrimental effect on memory. At the same time, COX-2 inhibitors engagement in the mechanisms of memory formation, and synapse plastic changes has been well documented. Our previous studies have demonstrated the contribution of cyclooxygenase-2 (COX-2) inhibition to the parameters of the mGluR5 pathway in memory formation. Because chronic administration of imipramine has been shown to affect mGluR5, the purpose of this study was to verify the hypothesis of COX-2 pathway engagement in disrupting effects of imipramine. Imipramine is currently used as a reference compound, and therefore it seems important to decipher and understand mood-related pathways, as well as cognitive changes activated during its use. This study covers the examination of spatial, and motor parameters. To this end, C57Bl/6J mice received imipramine, and NS398 (a COX-2 inhibitor) alone, or in combination for 7 or 14 days. We performed the modified Barnes maze (MBM), modified rotarod (MR) tests, and electrophysiological studies. The harmful effect of imipramine on MBM learning was improved by NS398 use. The same modulatory role of the COX-2 inhibitor in procedural learning in the MR test was found. In conclusion, our data show the involvement of the COX-2 pathway in changes in the long-term memory, and procedural memory of C57Bl/6J mice after chronic imipramine treatment.
Collapse
|
25
|
Djemil S, Chen X, Zhang Z, Lee J, Rauf M, Pak DTS, Dzakpasu R. Activation of nicotinic acetylcholine receptors induces potentiation and synchronization within in vitro hippocampal networks. J Neurochem 2019; 153:468-484. [PMID: 31821553 DOI: 10.1111/jnc.14938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are known to play a role in cognitive functions of the hippocampus, such as memory consolidation. Given that they conduct Ca2+ and are capable of regulating the release of glutamate and γ-aminobutyric acid (GABA) within the hippocampus, thereby shifting the excitatory-inhibitory ratio, we hypothesized that the activation of nAChRs will result in the potentiation of hippocampal networks and alter synchronization. We used nicotine as a tool to investigate the impact of activation of nAChRs on neuronal network dynamics in primary embryonic rat hippocampal cultures prepared from timed-pregnant Sprague-Dawley rats. We perturbed cultured hippocampal networks with increasing concentrations of bath-applied nicotine and performed network extracellular recordings of action potentials using a microelectrode array. We found that nicotine modulated network dynamics in a concentration-dependent manner; it enhanced firing of action potentials as well as facilitated bursting activity. In addition, we used pharmacological agents to determine the contributions of discrete nAChR subtypes to the observed network dynamics. We found that β4-containing nAChRs are necessary for the observed increases in spiking, bursting, and synchrony, while the activation of α7 nAChRs augments nicotine-mediated network potentiation but is not necessary for its manifestation. We also observed that antagonists of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs) partially blocked the effects of nicotine. Furthermore, nicotine exposure promoted autophosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII) and serine 831 phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit GluA1. These results suggest that nicotinic receptors induce potentiation and synchronization of hippocampal networks and glutamatergic synaptic transmission. Findings from this work highlight the impact of cholinergic signaling in generating network-wide potentiation in the form of enhanced spiking and bursting dynamics that coincide with molecular correlates of memory such as increased phosphorylation of CaMKII and GluA1. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Ziyue Zhang
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Jisoo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mikael Rauf
- Department of Human Science, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Rhonda Dzakpasu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Department of Physics, Georgetown University, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
26
|
Behavioral consequences of co-administration of MTEP and the COX-2 inhibitor NS398 in mice. Part 1. Behav Brain Res 2019; 370:111961. [DOI: 10.1016/j.bbr.2019.111961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/01/2023]
|
27
|
Weng QF, Chen GB, Xu MG, Long RT, Wang H, Wang XY, Jiang CN, Yi XN. Upregulation of PPAR-gamma activity inhibits cyclooxygenase 2 expression in cortical neurons with N-methyl-d-aspartic acid induced excitatory neurotoxicity. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1634488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Qi-Fang Weng
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Guo-Bin Chen
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Min-Guang Xu
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Ru-Tao Long
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Han Wang
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Xiao-Ying Wang
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Chao-Na Jiang
- Department of Physiology, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| | - Xi-Nan Yi
- Department of Human Anatomy, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, PR China
| |
Collapse
|
28
|
Panahi Y, Mojtahedzadeh M, Najafi A, Rajaee SM, Torkaman M, Sahebkar A. Neuroprotective Agents in the Intensive Care Unit: -Neuroprotective Agents in ICU. J Pharmacopuncture 2018; 21:226-240. [PMID: 30652049 PMCID: PMC6333194 DOI: 10.3831/kpi.2018.21.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/09/2018] [Accepted: 11/14/2018] [Indexed: 01/31/2023] Open
Abstract
Neuroprotection or prevention of neuronal loss is a complicated molecular process that is mediated by various cellular pathways. Use of different pharmacological agents as neuroprotectants has been reported especially in the last decades. These neuroprotective agents act through inhibition of inflammatory processes and apoptosis, attenuation of oxidative stress and reduction of free radicals. Control of this injurious molecular process is essential to the reduction of neuronal injuries and is associated with improved functional outcomes and recovery of the patients admitted to the intensive care unit. This study reviews neuroprotective agents and their mechanisms of action against central nervous system damages.
Collapse
Affiliation(s)
- Yunes Panahi
- Clinical Pharmacy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran,
Iran
- Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran,
Iran
| | - Mojtaba Mojtahedzadeh
- Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran,
Iran
- Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran,
Iran
| | - Atabak Najafi
- Gastrointestinal Pharmacology Interest Group(GPIG), Universal Scientific Education and Research Network(USERN), Tehran,
Iran
| | - Seyyed Mahdi Rajaee
- Gastrointestinal Pharmacology Interest Group(GPIG), Universal Scientific Education and Research Network(USERN), Tehran,
Iran
| | - Mohammad Torkaman
- Department of Pediatrics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran,
Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad,
Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad,
Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad,
Iran
| |
Collapse
|
29
|
Bazan NG. Docosanoids and elovanoids from omega-3 fatty acids are pro-homeostatic modulators of inflammatory responses, cell damage and neuroprotection. Mol Aspects Med 2018; 64:18-33. [PMID: 30244005 DOI: 10.1016/j.mam.2018.09.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
Abstract
The functional significance of the selective enrichment of the omega-3 essential fatty acid docosahexaenoic acid (DHA; 22C and 6 double bonds) in cellular membrane phospholipids of the nervous system is being clarified by defining its specific roles on membrane protein function and by the uncovering of the bioactive mediators, docosanoids and elovanoids (ELVs). Here, we describe the preferential uptake and DHA metabolism in photoreceptors and brain as well as the significance of the Adiponectin receptor 1 in DHA retention and photoreceptor cell (PRC) survival. We now know that this integral membrane protein is engaged in DHA retention as a necessary event for the function of PRCs and retinal pigment epithelial (RPE) cells. We present an overview of how a) NPD1 selectively mediates preconditioning rescue of RPE and PR cells; b) NPD1 restores aberrant neuronal networks in experimental epileptogenesis; c) the decreased ability to biosynthesize NPD1 in memory hippocampal areas of early stages of Alzheimer's disease takes place; d) NPD1 protection of dopaminergic circuits in an in vitro model using neurotoxins; and e) bioactivity elicited by DHA and NPD1 activate a neuroprotective gene-expression program that includes the expression of Bcl-2 family members affected by Aβ42, DHA, or NPD1. In addition, we highlight ELOVL4 (ELOngation of Very Long chain fatty acids-4), specifically the neurological and ophthalmological consequences of its mutations, and their role in providing precursors for the biosynthesis of ELVs. Then we outline evidence of ELVs ability to protect RPE cells, which sustain PRC integrity. In the last section, we present a summary of the protective bioactivity of docosanoids and ELVs in experimental ischemic stroke. The identification of early mechanisms of neural cell survival mediated by DHA-synthesized ELVs and docosanoids contributes to the understanding of cell function, pro-homeostatic cellular modulation, inflammatory responses, and innate immunity, opening avenues for prevention and therapeutic applications in neurotrauma, stroke and neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA.
| |
Collapse
|
30
|
Stark DT, Anderson DMG, Kwong JMK, Patterson NH, Schey KL, Caprioli RM, Caprioli J. Optic Nerve Regeneration After Crush Remodels the Injury Site: Molecular Insights From Imaging Mass Spectrometry. Invest Ophthalmol Vis Sci 2018; 59:212-222. [PMID: 29340649 PMCID: PMC5770179 DOI: 10.1167/iovs.17-22509] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Mammalian central nervous system axons fail to regenerate after injury. Contributing factors include limited intrinsic growth capacity and an inhibitory glial environment. Inflammation-induced optic nerve regeneration (IIR) is thought to boost retinal ganglion cell (RGC) intrinsic growth capacity through progrowth gene expression, but effects on the inhibitory glial environment of the optic nerve are unexplored. To investigate progrowth molecular changes associated with reactive gliosis during IIR, we developed an imaging mass spectrometry (IMS)-based approach that identifies discriminant molecular signals in and around optic nerve crush (ONC) sites. Methods ONC was performed in rats, and IIR was established by intravitreal injection of a yeast cell wall preparation. Optic nerves were collected at various postcrush intervals, and longitudinal sections were analyzed with matrix-assisted laser desorption/ionization (MALDI) IMS and data mining. Immunohistochemistry and confocal microscopy were used to compare discriminant molecular features with cellular features of reactive gliosis. Results IIR increased the area of the crush site that was occupied by a dense cellular infiltrate and mass spectral features consistent with lysosome-specific lipids. IIR also increased immunohistochemical labeling for microglia and macrophages. IIR enhanced clearance of lipid sulfatide myelin-associated inhibitors of axon growth and accumulation of simple GM3 gangliosides in a spatial distribution consistent with degradation of plasma membrane from degenerated axons. Conclusions IIR promotes a robust phagocytic response that improves clearance of myelin and axon debris. This growth-permissive molecular remodeling of the crush injury site extends our current understanding of IIR to include mechanisms extrinsic to the RGC.
Collapse
Affiliation(s)
- David T Stark
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - David M G Anderson
- Vanderbilt Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Jacky M K Kwong
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Nathan Heath Patterson
- Vanderbilt Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Kevin L Schey
- Vanderbilt Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Richard M Caprioli
- Vanderbilt Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Joseph Caprioli
- Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| |
Collapse
|
31
|
Li X, Sun W, An L. Nano-CuO impairs spatial cognition associated with inhibiting hippocampal long-term potentiation via affecting glutamatergic neurotransmission in rats. Toxicol Ind Health 2018; 34:409-421. [DOI: 10.1177/0748233718758233] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Manufactured metal nanoparticles and their applications are continuously expanding because of their unique characteristics while their increasing use may predispose to potential health problems. Several studies have reported the adverse effects of copper oxide nanoparticles (nano-CuO) relative to ecotoxicity and cell toxicity, whereas little is known about the neurotoxicity of nano-CuO. The present study aimed to examine its effects on spatial cognition, hippocampal function, and the possible mechanisms. Male Wistar rats were used to establish an animal model, and nano-CuO was administered at a dose of 0.5 mg/kg/day for 2 weeks. The Morris water maze (MWM) test was employed to evaluate learning and memory. The long-term potentiation (LTP) from Schaffer collaterals to the hippocampal CA1 region, and the effects of nano-CuO on synases were recorded in the hippocampal CA1 neurons of rats. MWM test showed that learning and memory abilities were impaired significantly by nano-CuO ( p < 0.05). The LTP test demonstrated that the field excitatory postsynaptic potential (fEPSP) slopes were significantly lower in nano-CuO-treated groups compared with the control group ( p < 0.01). Furthermore, the data of whole-cell patch-clamp experiments showed that nano-CuO markedly depressed the frequencies of both spontaneous excitatory postsynaptic currents (sEPSCs) and miniature EPSCs (mEPSCs), indicating an effect of nano-CuO on inhibiting the release frequency of glutamate presynapticly ( p < 0.01). Meanwhile, the amplitudes of both sEPSC and mEPSC were significantly reduced in nano-CuO-treated animals, which suggested that the effect of nano-CuO modulates postsynaptic receptor kinetics ( p < 0.01). Paired pulse facilitation (PPF) ( p < 0.05) and the expression of NR2A, but not NR2B, of N-methyl-d-aspartate (NMDA) subunits ( p < 0.05), were decreased significantly. In conclusion, nano-CuO impaired glutamate transmission presynapticly and postsynapticly, which may contribute importantly to diminished LTP and other induced cognitive deficits.
Collapse
Affiliation(s)
- Xiaoliang Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Sun
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei An
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Physiology, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
32
|
Retinal Pigment Epithelium and Photoreceptor Preconditioning Protection Requires Docosanoid Signaling. Cell Mol Neurobiol 2017; 38:901-917. [PMID: 29177613 PMCID: PMC5882642 DOI: 10.1007/s10571-017-0565-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023]
Abstract
Omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) are necessary for functional cell integrity. Preconditioning (PC), as we define it, is an acquired protection or resilience by a cell, tissue, or organ to a lethal stimulus enabled by a previous sublethal stressor or stimulus. In this study, we provide evidence that the omega-3 fatty acid docosahexaenoic acid (DHA) and its derivatives, the docosanoids 17-hydroxy docosahexaenoic acid (17-HDHA) and neuroprotectin D1 (NPD1), facilitate cell survival in both in vitro and in vivo models of retinal PC. We also demonstrate that PC requires the enzyme 15-lipoxygenase-1 (15-LOX-1), which synthesizes 17-HDHA and NPD1, and that this is specific to docosanoid signaling despite the concomitant release of the omega-6 arachidonic acid and eicosanoid synthesis. These findings advocate that DHA and docosanoids are protective enablers of PC in photoreceptor and retinal pigment epithelial cells.
Collapse
|
33
|
Bhattacharjee S, Jun B, Belayev L, Heap J, Kautzmann MA, Obenaus A, Menghani H, Marcell SJ, Khoutorova L, Yang R, Petasis NA, Bazan NG. Elovanoids are a novel class of homeostatic lipid mediators that protect neural cell integrity upon injury. SCIENCE ADVANCES 2017; 3:e1700735. [PMID: 28959727 PMCID: PMC5617374 DOI: 10.1126/sciadv.1700735] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/05/2017] [Indexed: 05/21/2023]
Abstract
We report the characterization of a novel class of lipid mediators termed elovanoids (ELVs) (ELV-N32 and ELV-N34), which are dihydroxylated derivatives of 32:6n3 and 34:6n3, respectively. The precursors of ELVs are made by elongation of a 22:6n3 fatty acid and catalyzed by ELOVL4 (elongation of very-long-chain fatty acids-4). The structure and stereochemistry of ELVs were established using synthetic compounds produced by stereocontrolled total synthesis. We report that ELV-mediated protection is induced in neuronal cultures undergoing either oxygen/glucose deprivation or N-methyl-d-aspartate receptor-mediated excitotoxicity, as well as in experimental ischemic stroke. The methyl ester or sodium salt of ELV-N32 and ELV-N34 resulted in reduced infarct volumes, promoted cell survival, and diminished neurovascular unit disruption when administered 1 hour following 2 hours of ischemia by middle cerebral artery occlusion. Together, our data reveal a novel prohomeostatic and neuroprotective lipid-signaling mechanism aiming to sustain neural cell integrity.
Collapse
Affiliation(s)
- Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ludmila Belayev
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jessica Heap
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Marie-Audrey Kautzmann
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Hemant Menghani
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Division of Hematology-Oncology, Department of Pediatrics, Louisiana State University Health Sciences Center and Children’s Hospital of New Orleans, New Orleans, LA 70118, USA
| | - Shawn J. Marcell
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Rong Yang
- Department of Chemistry, Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicos A. Petasis
- Department of Chemistry, Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicolas G. Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Corresponding author.
| |
Collapse
|
34
|
Asatryan A, Bazan NG. Molecular mechanisms of signaling via the docosanoid neuroprotectin D1 for cellular homeostasis and neuroprotection. J Biol Chem 2017; 292:12390-12397. [PMID: 28615451 PMCID: PMC5535015 DOI: 10.1074/jbc.r117.783076] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Docosahexaenoic acid, enriched in the brain and retina, generates docosanoids in response to disruptions of cellular homeostasis. Docosanoids include neuroprotectin D1 (NPD1), which is decreased in the CA1 hippocampal area of patients with early-stage Alzheimer's disease (AD). We summarize here how NPD1 elicits neuroprotection by up-regulating c-REL, a nuclear factor (NF)-κB subtype that, in turn, enhances expression of BIRC3 (baculoviral inhibitor of apoptosis repeat-containing protein 3) in the retina and in experimental stroke, leading to neuroprotection. Elucidating the mechanisms of action of docosanoids will contribute to managing diseases, including stroke, AD, age-related macular degeneration, traumatic brain injury, Parkinson's disease, and other neurodegenerations.
Collapse
Affiliation(s)
- Aram Asatryan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, Louisiana 70112-2223
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, Louisiana 70112-2223.
| |
Collapse
|
35
|
Elovanoids are novel cell-specific lipid mediators necessary for neuroprotective signaling for photoreceptor cell integrity. Sci Rep 2017; 7:5279. [PMID: 28706274 PMCID: PMC5509689 DOI: 10.1038/s41598-017-05433-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Docosahexaenoic acid (DHA, 22:6 n-3) is abundant in the retina and is enzymatically converted into pro-homeostatic docosanoids. The DHA- or eicosapentaenoic acid (EPA)-derived 26 carbon fatty acid is a substrate of elongase ELOVL4, which is expressed in photoreceptor cells and generates very long chain (≥C28) polyunsaturated fatty acids including n-3 (VLC-PUFAs,n-3). While ELOVL4 mutations are linked to vision loss and neuronal dysfunctions, the roles of VLC-PUFAs remain unknown. Here we report a novel class of lipid mediators biosynthesized in human retinal pigment epithelial (RPE) cells that are oxygenated derivatives of VLC-PUFAs,n-3; we termed these mediators elovanoids (ELV). ELVs have structures reminiscent of docosanoids but with different physicochemical properties and alternatively-regulated biosynthetic pathways. The structures, stereochemistry, and bioactivity of ELVs were determined using synthetic materials produced by stereo-controlled chemical synthesis. ELVs enhance expression of pro-survival proteins in cells undergoing uncompensated oxidative stress. Our findings unveil a novel autocrine/paracrine pro-homeostatic RPE cell signaling that aims to sustain photoreceptor cell integrity and reveal potential therapeutic targets for retinal degenerations.
Collapse
|
36
|
Sibarov DA, Abushik PA, Giniatullin R, Antonov SM. GluN2A Subunit-Containing NMDA Receptors Are the Preferential Neuronal Targets of Homocysteine. Front Cell Neurosci 2016; 10:246. [PMID: 27847466 PMCID: PMC5088185 DOI: 10.3389/fncel.2016.00246] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/07/2016] [Indexed: 12/30/2022] Open
Abstract
Homocysteine (HCY) is an endogenous redox active amino acid, best known as contributor to various neurodegenerative disorders. Although it is known that HCY can activate NMDA receptors (NMDARs), the mechanisms of its action on receptors composed of different NMDA receptor subunits remains almost unknown. In this study, using imaging and patch clamp technique in cultured cortical neurons and heterologous expression in HEK293T cells we tested the agonist activity of HCY on NMDARs composed of GluN1 and GluN2A subunits (GluN1/2A receptors) and GluN1 and GluN2B subunits (GluN1/2B receptors). We demonstrate that the time courses of Ca2+ transients and membrane currents activated by HCY and NMDA in cortical neurons are drastically different. Application of HCY to cortical neurons induced responses, which in contrast to currents induced by NMDA (both in the presence of glycine) considerably decreased to steady state of small amplitude. In contrast to NMDA, HCY-activated currents at steady state were resistant to the selective GluN2B subunit inhibitor ifenprodil. In calcium-free external solution the decrease of NMDA evoked currents was abolished, suggesting the Ca2+-dependent NMDAR desensitization. Under these conditions HCY evoked currents still declined almost to the baseline suggesting Ca2+-independent desensitization. In HEK293T cells HCY activated NMDARs of GluN1/2A and GluN1/2B subunit compositions with EC50s of 9.7 ± 1.8 and 61.8 ± 8.9 μM, respectively. Recombinant GluN1/2A receptors, however, did not desensitize by HCY, whereas GluN1/2B receptors were almost fully desensitized by HCY. Thus, HCY is a high affinity agonist of NMDARs preferring the GluN1/2A subunit composition. Our data suggest that HCY induced native NMDAR currents in neurons are mainly mediated by the "synaptic type" GluN1/2A NMDARs. This implies that in hyperhomocysteinemia, a disorder with enlarged level of HCY in plasma, HCY may persistently contribute to post-synaptic responses mediated by GluN2A-containing NMDA receptors. On the other hand, HCY toxicity may be limited by desensitization typical for HCY-induced activation of GluN2B-containing extrasynaptic receptors. Our findings, therefore, provide an evidence for the physiological relevance of endogenous HCY, which may represent an effective endogenous modulator of the central excitatory neurotransmission.
Collapse
Affiliation(s)
- Dmitry A Sibarov
- Laboratory of Comparative Neurophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences Saint-Petersburg, Russia
| | - Polina A Abushik
- Laboratory of Comparative Neurophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences Saint-Petersburg, Russia
| | - Rashid Giniatullin
- Department of Neurobiology, University of Eastern FinlandKuopio, Finland; Laboratory of Neurobiology, Kazan Federal UniversityKazan, Russia
| | - Sergei M Antonov
- Laboratory of Comparative Neurophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences Saint-Petersburg, Russia
| |
Collapse
|
37
|
Woodling NS, Colas D, Wang Q, Minhas P, Panchal M, Liang X, Mhatre SD, Brown H, Ko N, Zagol-Ikapitte I, van der Hart M, Khroyan TV, Chuluun B, Priyam PG, Milne GL, Rassoulpour A, Boutaud O, Manning-Boğ AB, Heller HC, Andreasson KI. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer's disease model mice. Brain 2016; 139:2063-81. [PMID: 27190010 DOI: 10.1093/brain/aww117] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-β accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-β oligomers.
Collapse
Affiliation(s)
- Nathaniel S Woodling
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 2 Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Damien Colas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qian Wang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paras Minhas
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maharshi Panchal
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xibin Liang
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Siddhita D Mhatre
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Holden Brown
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA 4 Brains On-line LLC, South San Francisco, CA, USA
| | - Novie Ko
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irene Zagol-Ikapitte
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marieke van der Hart
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taline V Khroyan
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bayarsaikhan Chuluun
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Prachi G Priyam
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ginger L Milne
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arash Rassoulpour
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Olivier Boutaud
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amy B Manning-Boğ
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Craig Heller
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin I Andreasson
- 1 Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
38
|
Bortolanza M, Padovan-Neto FE, Cavalcanti-Kiwiatkoski R, Dos Santos-Pereira M, Mitkovski M, Raisman-Vozari R, Del-Bel E. Are cyclooxygenase-2 and nitric oxide involved in the dyskinesia of Parkinson's disease induced by L-DOPA? Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0190. [PMID: 26009769 DOI: 10.1098/rstb.2014.0190] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Inflammatory mechanisms are proposed to play a role in L-DOPA-induced dyskinesia. Cyclooxygenase-2 (COX2) contributes to inflammation pathways in the periphery and is constitutively expressed in the central nervous system. Considering that inhibition of nitric oxide (NO) formation attenuates L-DOPA-induced dyskinesia, this study aimed at investigating if a NO synthase (NOS) inhibitor would change COX2 brain expression in animals with L-DOPA-induced dyskinesia. To this aim, male Wistar rats received unilateral 6-hydroxydopamine microinjection into the medial forebrain bundle were treated daily with L-DOPA (21 days) combined with 7-nitroindazole or vehicle. All hemi-Parkinsonian rats receiving l-DOPA showed dyskinesia. They also presented increased neuronal COX2 immunoreactivity in the dopamine-depleted dorsal striatum that was directly correlated with dyskinesia severity. Striatal COX2 co-localized with choline-acetyltransferase, calbindin and DARPP-32 (dopamine-cAMP-regulated phosphoprotein-32), neuronal markers of GABAergic neurons. NOS inhibition prevented L-DOPA-induced dyskinesia and COX2 increased expression in the dorsal striatum. These results suggest that increased COX2 expression after L-DOPA long-term treatment in Parkinsonian-like rats could contribute to the development of dyskinesia.
Collapse
Affiliation(s)
- Mariza Bortolanza
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil
| | - Fernando E Padovan-Neto
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil
| | - Roberta Cavalcanti-Kiwiatkoski
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Maurício Dos Santos-Pereira
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| | - Miso Mitkovski
- Light Microscopy Facility, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany
| | - Rita Raisman-Vozari
- Institut de Cerveau et de la Moelle Epinière, Sorbonne Université UPMC UM75 INSERM U1127, CNRS UMR 7225, Paris, France
| | - Elaine Del-Bel
- School of Odontology of Ribeirão Preto, Department of Morphology, University of São Paulo (USP), Physiology and Basic Pathology, Av. Café S/N, 14040-904, Ribeirão Preto, São Paulo, Brazil Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of Sao Paulo, São Paulo, Brazil Department of Behavioural Neurosciences, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, São Paulo, Brazil Medical School, Department of Physiology, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Saillet S, Quilichini PP, Ghestem A, Giusiano B, Ivanov AI, Hitziger S, Vanzetta I, Bernard C, Bénar CG. Interneurons contribute to the hemodynamic/metabolic response to epileptiform discharges. J Neurophysiol 2015; 115:1157-69. [PMID: 26745250 DOI: 10.1152/jn.00994.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 12/21/2015] [Indexed: 01/28/2023] Open
Abstract
Interpretation of hemodynamic responses in epilepsy is hampered by an incomplete understanding of the underlying neurovascular coupling, especially the contributions of excitation and inhibition. We made simultaneous multimodal recordings of local field potentials (LFPs), firing of individual neurons, blood flow, and oxygen level in the somatosensory cortex of anesthetized rats. Epileptiform discharges induced by bicuculline injections were used to trigger large local events. LFP and blood flow were robustly coupled, as were LFP and tissue oxygen. In a parametric linear model, LFP and the baseline activities of cerebral blood flow and tissue partial oxygen tension contributed significantly to blood flow and oxygen responses. In an analysis of recordings from 402 neurons, blood flow/tissue oxygen correlated with the discharge of putative interneurons but not of principal cells. Our results show that interneuron activity is important in the vascular and metabolic responses during epileptiform discharges.
Collapse
Affiliation(s)
- Sandrine Saillet
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France
| | - Pascale P Quilichini
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France
| | - Antoine Ghestem
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France
| | - Bernard Giusiano
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France; APHM, Timone Hospital, Division of Public Health, Marseille, France
| | - Anton I Ivanov
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France
| | | | - Ivo Vanzetta
- Aix-Marseille Université, CNRS, INT UMR 7289, Marseille, France
| | - Christophe Bernard
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France
| | - Christian-G Bénar
- INSERM, UMR 1106, Marseille, France; Aix-Marseille Université, Institut de Neurosciences des Systèmes, Marseille, France;
| |
Collapse
|
40
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015; 35:11791-810. [PMID: 26311764 DOI: 10.1523/jneurosci.0651-15.2015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
41
|
Patent highlights April–May 2015. Pharm Pat Anal 2015. [DOI: 10.4155/ppa.15.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
42
|
COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J Neurosci 2015. [PMID: 26311764 DOI: 10.1523/jneurosci.0651‐15.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Vasodilatory prostaglandins play a key role in neurovascular coupling (NVC), the tight link between neuronal activity and local cerebral blood flow, but their precise identity, cellular origin and the receptors involved remain unclear. Here we show in rats that NMDA-induced vasodilation and hemodynamic responses evoked by whisker stimulation involve cyclooxygenase-2 (COX-2) activity and activation of the prostaglandin E2 (PgE2) receptors EP2 and EP4. Using liquid chromatography-electrospray ionization-tandem mass spectrometry, we demonstrate that PgE2 is released by NMDA in cortical slices. The characterization of PgE2 producing cells by immunohistochemistry and single-cell reverse transcriptase-PCR revealed that pyramidal cells and not astrocytes are the main cell type equipped for PgE2 synthesis, one third expressing COX-2 systematically associated with a PgE2 synthase. Consistent with their central role in NVC, in vivo optogenetic stimulation of pyramidal cells evoked COX-2-dependent hyperemic responses in mice. These observations identify PgE2 as the main prostaglandin mediating sensory-evoked NVC, pyramidal cells as their principal source and vasodilatory EP2 and EP4 receptors as their targets. SIGNIFICANCE STATEMENT Brain function critically depends on a permanent spatiotemporal match between neuronal activity and blood supply, known as NVC. In the cerebral cortex, prostaglandins are major contributors to NVC. However, their biochemical identity remains elusive and their cellular origins are still under debate. Although astrocytes can induce vasodilations through the release of prostaglandins, the recruitment of this pathway during sensory stimulation is questioned. Using multidisciplinary approaches from single-cell reverse transcriptase-PCR, mass spectrometry, to ex vivo and in vivo pharmacology and optogenetics, we provide compelling evidence identifying PgE2 as the main prostaglandin in NVC, pyramidal neurons as their main cellular source and the vasodilatory EP2 and EP4 receptors as their main targets. These original findings will certainly change the current view of NVC.
Collapse
|
43
|
Zhou X, Chen Z, Yun W, Wang H. NMDA receptor activity determines neuronal fate: location or number? Rev Neurosci 2015; 26:39-47. [DOI: 10.1515/revneuro-2014-0053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/05/2014] [Indexed: 10/24/2022]
Abstract
AbstractIt is widely believed that the proper activation of N-methyl-D-aspartate (NMDA) receptors (NMDARs) promotes neuronal survival, whereas an excessive activation of NMDARs leads to neuronal damage. NMDARs are found at both synaptic and extrasynaptic sites. One current prevailing theory proposes the dichotomy of NMDAR activity. The role of the two population receptors is mutual antagonism. The activation of synaptic NMDARs, such as synaptic activity at physiological levels, promotes neuronal survival. However, the activation of extrasynaptic NMDARs occurring during stroke, brain injury, and chronic neurological diseases contributes to neuronal death. Thus, the location of NMDARs determines the neuronal fate. However, the theory is greatly challenged. Several studies suggested that synaptic NMDARs are involved in neuronal death. Recently, our work further showed that the coactivation of synaptic and extrasynaptic NMDARs contributes to neuronal death under neuronal insults. Therefore, we propose that the magnitude and duration of NMDAR activation determines the neuronal fate. More interestingly, there appears to be some subtle differences in the affinity between synaptic and extrasynaptic NMDARs, shedding light on the development of selective drugs to block extrasynaptic NMDARs.
Collapse
|
44
|
Bazan NG. Is there a molecular logic that sustains neuronal functional integrity and survival? Lipid signaling is necessary for neuroprotective neuronal transcriptional programs. Mol Neurobiol 2014; 50:1-5. [PMID: 25236258 DOI: 10.1007/s12035-014-8897-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
A challenge to civilization is the growing incidence in the loss of sight and cognition due to increased life expectancy. Therefore, we are confronted with a rise in the occurrence of photoreceptor- and neuronal-survival failure, as reflected mainly by age-related macular degeneration (AMD) and Alzheimer's disease (AD). Nervous system development is driven by neuronal apoptotic cell death, and thereafter, for the entire lifespan of an organism, neurons are postmitotic cells. In neurodegenerative diseases, apoptosis and other forms of cells death lead to selective neuronal loss. Although age is the main risk factor, not everyone develops these diseases during aging. Despite decades of important findings about neuronal cell death, the specific mechanisms that regulate neuronal survival remain incompletely understood.
Collapse
Affiliation(s)
- Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA,
| |
Collapse
|
45
|
Molokanova E, Akhtar MW, Sanz-Blasco S, Tu S, Piña-Crespo JC, McKercher SR, Lipton SA. Differential effects of synaptic and extrasynaptic NMDA receptors on Aβ-induced nitric oxide production in cerebrocortical neurons. J Neurosci 2014; 34:5023-8. [PMID: 24695719 PMCID: PMC3972726 DOI: 10.1523/jneurosci.2907-13.2014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 01/31/2014] [Accepted: 03/01/2014] [Indexed: 01/08/2023] Open
Abstract
Oligomerized amyloid-β (Aβ) peptide is thought to contribute to synaptic damage, resulting in dysfunctional neuronal networks in patients with Alzheimer's disease. It has been previously suggested that Aβ may be detrimental to neuronal health, at least in part, by triggering oxidative/nitrosative stress. However, the mechanisms underlying this process remain to be elucidated. Here, using rat primary cerebrocortical cultures, we demonstrate that Aβ1-42 oligomers trigger a dramatic increase in intracellular nitric oxide (NO) concentration via a process mediated by activation of NMDA-type glutamate receptors (NMDARs). Considering that synaptic NMDARs and extrasynaptic NMDARs (eNMDARs) can have opposite effects on neuronal viability, we explored their respective roles in Aβ-induced increases in NO levels. Surprisingly, after pharmacological isolation of eNMDARs, we discovered that eNMDARs are primarily responsible for the increase in neuronal NO triggered by Aβ oligomers. Moreover, we found that the eNMDAR-mediated increase in NO can produce S-nitrosylation of Drp1 (dynamin-related protein 1) and Cdk5 (cyclin-dependent kinase 5), targets known to contribute to Aβ-induced synaptic damage. These results suggest that pharmacological intervention specifically aimed at eNMDARs may decrease Aβ-induced nitrosative stress and thus ameliorate neurotoxic damage to synapses.
Collapse
Affiliation(s)
- Elena Molokanova
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Mohd Waseem Akhtar
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Sara Sanz-Blasco
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Shichun Tu
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Juan C. Piña-Crespo
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Scott R. McKercher
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Stuart A. Lipton
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
46
|
Liao D, Miller EC, Teravskis PJ. Tau acts as a mediator for Alzheimer's disease-related synaptic deficits. Eur J Neurosci 2014; 39:1202-13. [PMID: 24712999 PMCID: PMC3983570 DOI: 10.1111/ejn.12504] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/04/2014] [Accepted: 01/06/2014] [Indexed: 12/11/2022]
Abstract
The two histopathological hallmarks of Alzheimer's disease (AD) are amyloid plaques containing multiple forms of amyloid beta (Aβ) and neurofibrillary tangles containing phosphorylated tau proteins. As mild cognitive impairment frequently occurs long before the clinical diagnosis of AD, the scientific community has been increasingly interested in the roles of Aβ and tau in earlier cellular changes that lead to functional deficits. Therefore, great progress has recently been made in understanding how Aβ or tau causes synaptic dysfunction. However, the interaction between the Aβ and tau-initiated intracellular cascades that lead to synaptic dysfunction remains elusive. The cornerstone of the two-decade-old hypothetical amyloid cascade model is that amyloid pathologies precede tau pathologies. Although the premise of Aβ-tau pathway remains valid, the model keeps evolving as new signaling events are discovered that lead to functional deficits and neurodegeneration. Recent progress has been made in understanding Aβ-PrP(C) -Fyn-mediated neurotoxicity and synaptic deficits. Although still elusive, many novel upstream and downstream signaling molecules have been found to modulate tau mislocalization and tau hyperphosphorylation. Here we will discuss the mechanistic interactions between Aβ-PrP(C) -mediated neurotoxicity and tau-mediated synaptic deficits in an updated amyloid cascade model with calcium and tau as the central mediators.
Collapse
Affiliation(s)
- Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455
| | - Eric C. Miller
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455
| | - Peter J. Teravskis
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455
- College of Biological Sciences University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
47
|
Kaiser O, Aliuos P, Wissel K, Lenarz T, Werner D, Reuter G, Kral A, Warnecke A. Dissociated neurons and glial cells derived from rat inferior colliculi after digestion with papain. PLoS One 2013; 8:e80490. [PMID: 24349001 PMCID: PMC3861243 DOI: 10.1371/journal.pone.0080490] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/13/2013] [Indexed: 01/10/2023] Open
Abstract
The formation of gliosis around implant electrodes for deep brain stimulation impairs electrode–tissue interaction. Unspecific growth of glial tissue around the electrodes can be hindered by altering physicochemical material properties. However, in vitro screening of neural tissue–material interaction requires an adequate cell culture system. No adequate model for cells dissociated from the inferior colliculus (IC) has been described and was thus the aim of this study. Therefore, IC were isolated from neonatal rats (P3_5) and a dissociated cell culture was established. In screening experiments using four dissociation methods (Neural Tissue Dissociation Kit [NTDK] T, NTDK P; NTDK PN, and a validated protocol for the dissociation of spiral ganglion neurons [SGN]), the optimal media, and seeding densities were identified. Thereafter, a dissociation protocol containing only the proteolytic enzymes of interest (trypsin or papain) was tested. For analysis, cells were fixed and immunolabeled using glial- and neuron-specific antibodies. Adhesion and survival of dissociated neurons and glial cells isolated from the IC were demonstrated in all experimental settings. Hence, preservation of type-specific cytoarchitecture with sufficient neuronal networks only occurred in cultures dissociated with NTDK P, NTDK PN, and fresh prepared papain solution. However, cultures obtained after dissociation with papain, seeded at a density of 2×104 cells/well and cultivated with Neuro Medium for 6 days reliably revealed the highest neuronal yield with excellent cytoarchitecture of neurons and glial cells. The herein described dissociated culture can be utilized as in vitro model to screen interactions between cells of the IC and surface modifications of the electrode.
Collapse
Affiliation(s)
- Odett Kaiser
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Pooyan Aliuos
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Kirsten Wissel
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Darja Werner
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Günter Reuter
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Andrej Kral
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
48
|
Siddoway B, Hou H, Yang H, Petralia R, Xia H. Synaptic activity bidirectionally regulates a novel sequence-specific S-Q phosphoproteome in neurons. J Neurochem 2013; 128:841-51. [PMID: 24117848 DOI: 10.1111/jnc.12487] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 12/01/2022]
Abstract
Protein phosphorylation plays a critical role in neuronal transcription, translation, cell viability, and synaptic plasticity. In neurons, phospho-enzymes and specific substrates directly link glutamate release and post-synaptic depolarization to these cellular functions; however, many of these enzymes and their protein substrates remain uncharacterized or unidentified. In this article, we identify a novel, synaptically driven neuronal phosphoproteome characterized by a specific motif of serine/threonine-glutamine ([S/T]-Q, abbreviated as SQ). These SQ-containing substrates are predominantly localized to dendrites, synapses, the soma; and activation of this SQ phosphoproteome by bicuculline application is induced via calcium influx through L-type calcium channels. On the other hand, acute application of NMDA can inactivate this SQ phosphoproteome. We demonstrate that the SQ motif kinase Ataxia-telangiectasia mutated can also localize to dendrites and dendritic spines, in addition to other subcellular compartments, and is activated by bicuculline application. Pharmacology studies indicate that Ataxia-telangiectasia mutated and its sister kinase ataxia telangiectasia mutated and Rad3-related up-regulate these neuronal SQ substrates. Phosphoproteomics identified over 150 SQ-containing substrates whose phosphorylation is bidirectionally regulated by synaptic activity.
Collapse
Affiliation(s)
- Benjamin Siddoway
- Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
49
|
van Leyen K. Lipoxygenase: an emerging target for stroke therapy. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2013; 12:191-9. [PMID: 23394536 DOI: 10.2174/18715273112119990053] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Revised: 08/08/2012] [Accepted: 08/21/2012] [Indexed: 02/06/2023]
Abstract
Neuroprotection as approach to stroke therapy has recently seen a revival of sorts, fueled in part by the continuing necessity to improve acute stroke care, and in part by the identification of novel drug targets. 12/15- Lipoxygenase (12/15-LOX), one of the key enzymes of the arachidonic acid cascade, contributes to both neuronal cell death and vascular injury. Inhibition of 12/15-LOX may thus provide multifactorial protection against ischemic injury. Targeting 12/15-LOX and related eicosanoid pathways is the subject of this brief review.
Collapse
Affiliation(s)
- Klaus van Leyen
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, 149 13th St., R. 2401, Charlestown, MA 02129, USA.
| |
Collapse
|
50
|
Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflammation: the role and consequences. Neurosci Res 2013; 79:1-12. [PMID: 24144733 DOI: 10.1016/j.neures.2013.10.004] [Citation(s) in RCA: 491] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/07/2013] [Accepted: 10/09/2013] [Indexed: 01/05/2023]
Abstract
Neuroinflammation is central to the common pathology of several acute and chronic brain diseases. This review examines the consequences of excessive and prolonged neuroinflammation, particularly its damaging effects on cellular and/or brain function, as well as its relevance to disease progression and possible interventions. The evidence gathered here indicates that neuroinflammation causes and accelerates long-term neurodegenerative disease, playing a central role in the very early development of chronic conditions including dementia. The wide scope and numerous complexities of neuroinflammation suggest that combinations of different preventative and therapeutic approaches may be efficacious.
Collapse
Affiliation(s)
- Monty Lyman
- Section of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK
| | - Dafydd G Lloyd
- Section of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Marcela P Vizcaychipi
- Section of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK
| | - Daqing Ma
- Section of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK.
| |
Collapse
|