1
|
Depressive-like behaviors induced by chronic cerebral hypoperfusion associate with a dynamic change of GABA B1/B2 receptors expression in hippocampal CA1 region. Physiol Behav 2022; 254:113887. [PMID: 35724927 DOI: 10.1016/j.physbeh.2022.113887] [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: 03/07/2022] [Revised: 05/07/2022] [Accepted: 06/16/2022] [Indexed: 11/21/2022]
Abstract
Cerebral ischemia could induce depressive-like behaviors; however, the alteration of gamma-aminobutyric acid receptors type B (GABAB) receptors in these pathological processes has not been extensively investigated. The aim of the current study was to document the behavioral change and the alteration of GABAB receptors in chronic cerebral hypoperfusion. The permanent occlusion of the bilateral common carotid arteries (two-vessel occlusion, 2VO) was performed to induce chronic cerebral ischemia (CCH). The depressive-like behaviors were evaluated with sucrose preference test, novelty suppress feeding test as well as forced swim test at 4, 8, and 12 weeks after the 2VO surgery. The total, surface and intracellular expressions of GABAB subunit 1 (GABAB1) and subunit 2 (GABAB2) in hippocampal CA1 were quantified by western blot. The depressive-like behaviors were observed in rats suffered from 4, 8, and 12 weeks 2VO in sucrose preference test, novelty suppress feeding test and forced swim test. In addition, the surface and total expression of GABAB1 in CA1 was reduced at 4 weeks after 2VO rather than 8 or 12 weeks. While the surface and total expression of GABAB2 in CA1 was decreased throughout the ischemia timeline (4, 8, and 12 weeks). Taken together, our findings suggested the potential roles of GABAB1 and GABAB2 subunits involved in depressive-like behaviors caused by chronic cerebral hypoperfusion.
Collapse
|
2
|
Balakrishnan K, Hleihil M, Bhat MA, Ganley RP, Vaas M, Klohs J, Zeilhofer HU, Benke D. Targeting the interaction of GABA B receptors with CaMKII with an interfering peptide restores receptor expression after cerebral ischemia and inhibits progressive neuronal death in mouse brain cells and slices. Brain Pathol 2022; 33:e13099. [PMID: 35698024 PMCID: PMC9836377 DOI: 10.1111/bpa.13099] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/29/2022] [Indexed: 01/21/2023] Open
Abstract
Cerebral ischemia is the leading cause for long-term disability and mortality in adults due to massive neuronal death. Currently, there is no pharmacological treatment available to limit progressive neuronal death after stroke. A major mechanism causing ischemia-induced neuronal death is the excessive release of glutamate and the associated overexcitation of neurons (excitotoxicity). Normally, GABAB receptors control neuronal excitability in the brain via prolonged inhibition. However, excitotoxic conditions rapidly downregulate GABAB receptors via a CaMKII-mediated mechanism and thereby diminish adequate inhibition that could counteract neuronal overexcitation and neuronal death. To prevent the deleterious downregulation of GABAB receptors, we developed a cell-penetrating synthetic peptide (R1-Pep) that inhibits the interaction of GABAB receptors with CaMKII. Administration of this peptide to cultured cortical neurons exposed to excitotoxic conditions restored cell surface expression and function of GABAB receptors. R1-Pep did not affect CaMKII expression or activity but prevented its T286 autophosphorylation that renders it autonomously and persistently active. Moreover, R1-Pep counteracted the aberrant downregulation of G protein-coupled inwardly rectifying K+ channels and the upregulation of N-type voltage-gated Ca2+ channels, the main effectors of GABAB receptors. The restoration of GABAB receptors activated the Akt survival pathway and inhibited excitotoxic neuronal death with a wide time window in cultured neurons. Restoration of GABAB receptors and neuroprotective activity of R1-Pep was verified by using brain slices prepared from mice after middle cerebral artery occlusion (MCAO). Treatment with R1-Pep restored normal GABAB receptor expression and GABA receptor-mediated K+ channel currents. This reduced MCAO-induced neuronal excitability and inhibited neuronal death. These results support the hypothesis that restoration of GABAB receptor expression under excitatory conditions provides neuroprotection and might be the basis for the development of a selective intervention to inhibit progressive neuronal death after ischemic stroke.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Present address:
Dewpoint Therapeutics GMBHDresdenGermany
| | - Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Musadiq A. Bhat
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland
| | - Robert P. Ganley
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland
| | - Markus Vaas
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Present address:
Clinical Trial Center ZurichUniversity Hospital of ZurichZurichSwitzerland
| | - Jan Klohs
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Institute for Biomedical Engineering, University of Zurich and ETH ZurichZurichSwitzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Drug Discovery Network ZurichZurichSwitzerland,Institute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Drug Discovery Network ZurichZurichSwitzerland
| |
Collapse
|
3
|
GSK-126 Protects CA1 Neurons from H3K27me3-Mediated Apoptosis in Cerebral Ischemia. Mol Neurobiol 2022; 59:2552-2562. [PMID: 35091962 PMCID: PMC9016005 DOI: 10.1007/s12035-021-02677-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
Epigenetics, including histone modifications, play a significant role in central nervous system diseases, but the underlying mechanism remains to be elucidated. The aim of this study was to evaluate the role of H3K27me3 in regulating transcriptomic and pathogenic mechanisms following global ischemic stroke. Here, we found that in vivo ischemic/reperfusion (I/R) injury induced marked upregulation of H3K27me3 in the hippocampus. The administration of GSK-126 to rat brains decreased the levels of H3K27me3 in the hippocampus and reduced neuronal apoptosis after experimental stroke. Furthermore, ChIP-seq data demonstrated that the primary role of GSK-126 in the ischemic brain is to reduce H3K27me3 enrichment, mediating negative regulation of the execution phase of apoptosis and the MAPK signaling pathway. Further study suggested that the protective role of GSK-126 in ischemic rats was antagonized by U0126, an inhibitor of ERK1/2. Collectively, we demonstrated the potential of H3K27me3 as a novel stroke therapeutic target, and GSK-126 exerted a neuroprotective function in ischemic brain injury, which might be associated with activation of the MAPK/ERK pathway.
Collapse
|
4
|
Hleihil M, Vaas M, Bhat MA, Balakrishnan K, Benke D. Sustained Baclofen-Induced Activation of GABA B Receptors After Cerebral Ischemia Restores Receptor Expression and Function and Limits Progressing Loss of Neurons. Front Mol Neurosci 2021; 14:726133. [PMID: 34539344 PMCID: PMC8440977 DOI: 10.3389/fnmol.2021.726133] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
One important function of GABAB receptors is the control of neuronal activity to prevent overexcitation and thereby excitotoxic death, which is a hallmark of cerebral ischemia. Consequently, sustained activation of GABAB receptors with the selective agonist baclofen provides neuroprotection in in vitro and in vivo models of cerebral ischemia. However, excitotoxic conditions severely downregulate the receptors, which would compromise the neuroprotective effectiveness of baclofen. On the other hand, recent work suggests that sustained activation of GABAB receptors stabilizes receptor expression. Therefore, we addressed the question whether sustained activation of GABAB receptors reduces downregulation of the receptor under excitotoxic conditions and thereby preserves GABAB receptor-mediated inhibition. In cultured neurons subjected to oxygen and glucose deprivation (OGD), to mimic cerebral ischemia, GABAB receptors were severely downregulated. Treatment of the cultures with baclofen after OGD restored GABAB receptor expression and reduced loss of neurons. Restoration of GABAB receptors was due to enhanced fast recycling of the receptors, which reduced OGD-induced sorting of the receptors to lysosomal degradation. Utilizing the middle cerebral artery occlusion (MCAO) mouse model of cerebral ischemia, we verified the severe downregulation of GABAB receptors in the affected cortex and a partial restoration of the receptors after systemic injection of baclofen. Restored receptor expression recovered GABAB receptor-mediated currents, normalized the enhanced neuronal excitability observed after MCAO and limited progressive loss of neurons. These results suggest that baclofen-induced restoration of GABAB receptors provides the basis for the neuroprotective activity of baclofen after an ischemic insult. Since GABAB receptors regulate multiple beneficial pathways, they are promising targets for a neuroprotective strategy in acute cerebral ischemia.
Collapse
Affiliation(s)
- Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Markus Vaas
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Musadiq A Bhat
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Vahidinia Z, Mahdavi E, Talaei SA, Naderian H, Tamtaji A, Haddad Kashani H, Beyer C, Azami Tameh A. The effect of female sex hormones on Hsp27 phosphorylation and histological changes in prefrontal cortex after tMCAO. Pathol Res Pract 2021; 221:153415. [PMID: 33857717 DOI: 10.1016/j.prp.2021.153415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Female sex hormones are protective factors against many neurological disorders such as brain ischemia. Heat shock protein like HSP27 is activated after tissue injury. The main purpose of the present study is to determine the effect of a combined estrogen / progesterone cocktail on the morphology of astrocytes, neurons and Hsp27 phosphorylation after cerebral ischemia. METHODS One hour after the MCAO induction, a single dose of estrogen and progesterone was injected. The infarct volume was calculated by TTC staining 24 h after ischemia. Immunohistochemistry was used to show the effects of estrogen and progesterone on astrocyte and neuron morphology, as well as the Western blot technique used for the quantitation of phosphorylated Hsp27. RESULTS The combined dose of estrogen and progesterone significantly decreased astrocytosis after ischemia and increased neuron survival. There was a large increase in Hsp27 phosphorylation in the penumbra ischemic region after stroke, which was significantly reduced by hormone therapy. CONCLUSION Our results indicate that the neuroprotective effect of neurosteroids in the brain may be due to the modulation of heat shock proteins.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Elham Mahdavi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Homayoun Naderian
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Aboutaleb Tamtaji
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Haddad Kashani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Tabary M, Aryannejad A, Noroozi N, Tavangar SM, Mohammad Jafari R, Araghi F, Dadkhahfar S, Dehpour AR. Ivermectin Increases Random-Pattern Skin Flap Survival in Rats: The Novel Role of GABAergic System. J Surg Res 2021; 259:431-441. [PMID: 33069391 DOI: 10.1016/j.jss.2020.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ivermectin (IVM) was first used as an antiparasitic agent; however, the role of this drug evolved into a broad spectrum. Many mechanisms have been proposed, including interaction with the GABAergic system. Considering the presence of GABA receptor in the skin tissue and its role in ischemia-reperfusion I/R injury, we aimed to evaluate the effect of IVM through GABA receptors on random-pattern skin flap survival. METHODS Sixty Wistar male rats were used. Multiple doses of IVM (0.01, 0.05, 0.2, and 0.5 mg/kg) were injected intraperitoneally before the surgery. Baclofen (selective GABAB agonist) and bicuculline (selective GABAA antagonist) were administered in combination with IVM to assess the role of the GABAergic system. Histopathological evaluations, immunohistochemical staining, quantitative assessment of IL-1β and TNFα, and the expression of GABAA α1 subunit and GABAB R1 receptors were evaluated in the skin tissue. RESULTS IVM 0.05 mg/kg could significantly increase flap survival compared with the control group (P < 0.001). Subeffective dose of baclofen (0.1 mg/kg) had synergistic effect with the subeffective dose of IVM (0.01 mg/kg) (P < 0.001), whereas bicuculline 1 mg/kg reversed the effect of IVM (0.05 mg/kg) (P < 0.001). IVM 0.05 mg/kg could also decrease the IL-1β and TNFα levels and increase the expression of GABAA α1 subunit and GABAB R1 receptors in the flap tissue compared with the control group. CONCLUSIONS IVM could improve skin flap survival, probably mediated by the GABAergic pathway. Both GABAA and GABAB receptors are involved in this process. This finding may repurpose the use of old drug, "Ivermectin."
Collapse
Affiliation(s)
- Mohammadreza Tabary
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Armin Aryannejad
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafise Noroozi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Araghi
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Dadkhahfar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
The Roles of GABA in Ischemia-Reperfusion Injury in the Central Nervous System and Peripheral Organs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4028394. [PMID: 31814874 PMCID: PMC6878816 DOI: 10.1155/2019/4028394] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 10/18/2019] [Indexed: 12/30/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a common pathological process, which may lead to dysfunctions and failures of multiple organs. A flawless medical way of endogenous therapeutic target can illuminate accurate clinical applications. γ-Aminobutyric acid (GABA) has been known as a marker in I/R injury of the central nervous system (mainly in the brain) for a long time, and it may play a vital role in the occurrence of I/R injury. It has been observed that throughout cerebral I/R, levels, syntheses, releases, metabolisms, receptors, and transmissions of GABA undergo complex pathological variations. Scientists have investigated the GABAergic enhancers for attenuating cerebral I/R injury; however, discussions on existing problems and mechanisms of available drugs were seldom carried out so far. Therefore, this review would summarize the process of pathological variations in the GABA system under cerebral I/R injury and will cover corresponding probable issues and mechanisms in using GABA-related drugs to illuminate the concern about clinical illness for accurately preventing cerebral I/R injury. In addition, the study will summarize the increasing GABA signals that can prevent I/R injuries occurring in peripheral organs, and the roles of GABA were also discussed correspondingly.
Collapse
|
8
|
da Silveira TL, Machado ML, Arantes LP, Zamberlan DC, Cordeiro LM, Obetine FBB, da Silva AF, Tassi CL, Soares FAA. Guanosine Prevents against Glutamatergic Excitotoxicity in C. elegans. Neuroscience 2019; 414:265-272. [PMID: 31306683 DOI: 10.1016/j.neuroscience.2019.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
Glutamatergic neurotransmission is present in most mammalian excitatory synapses and plays a key role in central nervous system homeostasis. When over-activated, it can induce excitotoxicity, which is present in several neuropathologies. The nucleoside guanosine (GUO) is a guanine-based purine known to have neuroprotective effects by modulating glutamatergic system during glutamate excitotoxicity in mammals. However, GUO action in Caenorhabditis elegans, as well as on C. elegans glutamatergic excitotoxicity model, is not known. The GUO effects on behavioral parameters in Wild Type (WT) and knockouts worms for glutamate transporters (GLT-3, GLT-1), glutamate vesicular transporter (EAT-4), and NMDA and non-NMDA receptors were used to evaluate the GUO modulatory effects. The GUO tested concentrations did not alter the animals' development, but GUO reduced pharyngeal pumps in WT animals in a dose-dependent manner. The same effect was observed in pharyngeal pumps, when the animals were treated with 4 mM of GUO in glr-1, nmr-1 and eat-4, but not in glt-3 and glt-3;glt-1 knockouts. The double mutant glt-3; glt-1 for GluTs had decreased body bends and an increased number of reversions. This effect was reverted after treatment with GUO. Furthermore, GUO did not alter the sensory response in worms with altered glutamatergic signaling. Thus, GUO seems to modulate the worm's glutamatergic system in situations of exacerbated glutamatergic signaling, which are represented by knockout strains to glutamate transporters. However, in WT animals, GUO appears to reinforce glutamatergic signaling in specific neurons. Our findings indicate that C. elegans strains are useful models to study new compounds that could be used in glutamate-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Tássia Limana da Silveira
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Marina Lopes Machado
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Leticia Priscilla Arantes
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Daniele Coradini Zamberlan
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Larissa Marafiga Cordeiro
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Fabiane Bicca Baptista Obetine
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Aline Franzen da Silva
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cintia Letícia Tassi
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Felix Alexandre Antunes Soares
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
9
|
Ruan L, Wang Y, Chen SC, Zhao T, Huang Q, Hu ZL, Xia NZ, Liu JJ, Chen WJ, Zhang Y, Cheng JL, Gao HC, Yang YJ, Sun HZ. Metabolite changes in the ipsilateral and contralateral cerebral hemispheres in rats with middle cerebral artery occlusion. Neural Regen Res 2017; 12:931-937. [PMID: 28761426 PMCID: PMC5514868 DOI: 10.4103/1673-5374.208575] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cerebral ischemia not only causes pathological changes in the ischemic areas but also induces a series of secondary changes in more distal brain regions (such as the contralateral cerebral hemisphere). The impact of supratentorial lesions, which are the most common type of lesion, on the contralateral cerebellum has been studied in patients by positron emission tomography, single photon emission computed tomography, magnetic resonance imaging and diffusion tensor imaging. In the present study, we investigated metabolite changes in the contralateral cerebral hemisphere after supratentorial unilateral ischemia using nuclear magnetic resonance spectroscopy-based metabonomics. The permanent middle cerebral artery occlusion model of ischemic stroke was established in rats. Rats were randomly divided into the middle cerebral artery occlusion 1-, 3-, 9- and 24-hour groups and the sham group. 1H nuclear magnetic resonance spectroscopy was used to detect metabolites in the left and right cerebral hemispheres. Compared with the sham group, the concentrations of lactate, alanine, γ-aminobutyric acid, choline and glycine in the ischemic cerebral hemisphere were increased in the acute stage, while the concentrations of N-acetyl aspartate, creatinine, glutamate and aspartate were decreased. This demonstrates that there is an upregulation of anaerobic glycolysis (shown by the increase in lactate), a perturbation of choline metabolism (suggested by the increase in choline), neuronal cell damage (shown by the decrease in N-acetyl aspartate) and neurotransmitter imbalance (evidenced by the increase in γ-aminobutyric acid and glycine and by the decrease in glutamate and aspartate) in the acute stage of cerebral ischemia. In the contralateral hemisphere, the concentrations of lactate, alanine, glycine, choline and aspartate were increased, while the concentrations of γ-aminobutyric acid, glutamate and creatinine were decreased. This suggests that there is a difference in the metabolite changes induced by ischemic injury in the contralateral and ipsilateral cerebral hemispheres. Our findings demonstrate the presence of characteristic changes in metabolites in the contralateral hemisphere and suggest that they are most likely caused by metabolic changes in the ischemic hemisphere.
Collapse
Affiliation(s)
- Lei Ruan
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yan Wang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shu-Chao Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tian Zhao
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qun Huang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zi-Long Hu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Neng-Zhi Xia
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jin-Jin Liu
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wei-Jian Chen
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yong Zhang
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing-Liang Cheng
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hong-Chang Gao
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yun-Jun Yang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hou-Zhang Sun
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
10
|
Glial GABA Transporters as Modulators of Inhibitory Signalling in Epilepsy and Stroke. ADVANCES IN NEUROBIOLOGY 2017; 16:137-167. [PMID: 28828609 DOI: 10.1007/978-3-319-55769-4_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Imbalances in GABA-mediated tonic inhibition are involved in several pathophysiological conditions. A classical way of controlling tonic inhibition is through pharmacological intervention with extrasynaptic GABAA receptors that sense ambient GABA and mediate a persistent GABAergic conductance. An increase in tonic inhibition may, however, also be obtained indirectly by inhibiting glial GABA transporters (GATs). These are sodium-coupled membrane transport proteins that normally act to terminate GABA neurotransmitter action by taking up GABA into surrounding astrocytes. The aim of the review is to provide an overview of glial GATs in regulating tonic inhibition, especially in epilepsy and stroke. This entails a comprehensive summary of changes known to occur in GAT expression levels and signalling following epileptic and ischemic insults. Further, we discuss the accumulating pharmacological evidence for targeting GATs in these diseases.
Collapse
|
11
|
Vahidinia Z, Alipour N, Atlasi MA, Naderian H, Beyer C, Azami Tameh A. Gonadal steroids block the calpain-1-dependent intrinsic pathway of apoptosis in an experimental rat stroke model. Neurol Res 2016; 39:54-64. [PMID: 27832728 DOI: 10.1080/01616412.2016.1250459] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Apoptosis plays an important role in the progression of the ischemic penumbra after reperfusion. Estrogen and progesterone have neuroprotective effects against ischemic brain damage, however the exact mechanisms of neuroprotection and signaling pathways is not completely understood. In this study, we investigated the possible regulatory effects of a combined steroid treatment on extrinsic and intrinsic apoptotic signaling pathways after cerebral ischemia. METHODS Adult male Wistar rats were subjected to transient middle cerebral artery occlusion (tMCAO) using an intraluminal filament technique for 1 h followed by 23 h reperfusion. Estrogen and progesterone were immediately injected after tMCAO subcutaneously. Sensorimotor functional tests and the infarct volume were evaluated 24 h after ischemia. Protein expression of calpain-1 and Fas receptor (FasR), key members of intrinsic and extrinsic apoptosis, were determined in the penumbra region of the ischemic brain using western blot analysis, immunohistochemistry, and TUNEL staining. RESULTS Neurological deficits and infarct volume were significantly reduced following hormone therapy. Calpain-1 up-regulation and caspase-3 activation were apparent 24 h after ischemia in the peri-infarct area of the cerebral cortex. Steroid hormone treatment reduced infarct pathology and attenuated the induction of both proteases. FasR protein levels were not affected by ischemia and hormone application. CONCLUSION We conclude that a combined steroid treatment inhibits ischemia-induced neuronal apoptosis through the regulation of intrinsic pathways.
Collapse
Key Words
- Apoptosis
- CBF, Cerebral blood flow
- CCA, Common carotid artery
- CNS, Central nervous system
- Calpain-1
- DISC, Death inducing signaling complex
- Estrogen
- FasR, Fas receptor
- GFAP, Glial fibrillary acidic protein
- HRP, Horseradish peroxidase
- I/R, Ischemia/reperfusion
- ICA, Internal carotid artery
- IHC, Immunohistochemistry
- MCA, Middle cerebral artery
- MCAO, Middle cerebral artery occlusion
- NeuN, Neuronal nuclear antigen
- PBS, Phosphate-buffered saline
- PU, Perfusion units
- PVDF, Polyvinylidene fluoride
- Progesterone
- RIPA, Radioimmunoprecipitation assay
- ROS, Reactive oxygen species
- SDS, Sodium dodecyl sulfate
- TBI, Traumatic brain ischemia
- TNF, Tumor necrosis factor
- TTC, Triphenyltetrazolium chloride
- TUNEL, Terminal deoxynucleotidyltransferase (TdT)-mediated dUTP-biotin nick-end labeling
- tMCAO
- tMCAO, transient middle cerebral artery occlusion
Collapse
Affiliation(s)
- Zeinab Vahidinia
- a Anatomical Sciences Research Center , Kashan University of Medical Sciences , Kashan , Iran
| | - Nasim Alipour
- a Anatomical Sciences Research Center , Kashan University of Medical Sciences , Kashan , Iran
| | - Mohammad Ali Atlasi
- a Anatomical Sciences Research Center , Kashan University of Medical Sciences , Kashan , Iran
| | - Homayoun Naderian
- a Anatomical Sciences Research Center , Kashan University of Medical Sciences , Kashan , Iran
| | - Cordian Beyer
- b Faculty of Medicine , Institute of Neuroanatomy, RWTH Aachen University , Aachen , Germany
| | - Abolfazl Azami Tameh
- a Anatomical Sciences Research Center , Kashan University of Medical Sciences , Kashan , Iran.,c Department of Anatomy, School of Medicine , Kashan University of Medical Sciences , Kashan , Iran
| |
Collapse
|
12
|
Alleviation of glutamate mediated neuronal insult by piroxicam in rodent model of focal cerebral ischemia: a possible mechanism of GABA agonism. J Physiol Biochem 2014; 70:901-13. [DOI: 10.1007/s13105-014-0358-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/17/2014] [Indexed: 12/17/2022]
|
13
|
Nuñez-Figueredo Y, Ramírez-Sánchez J, Delgado-Hernández R, Porto-Verdecia M, Ochoa-Rodríguez E, Verdecia-Reyes Y, Marin-Prida J, González-Durruthy M, Uyemura SA, Rodrigues FP, Curti C, Souza DO, Pardo-Andreu GL. JM-20, a novel benzodiazepine–dihydropyridine hybrid molecule, protects mitochondria and prevents ischemic insult-mediated neural cell death in vitro. Eur J Pharmacol 2014; 726:57-65. [DOI: 10.1016/j.ejphar.2014.01.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 12/21/2013] [Accepted: 01/07/2014] [Indexed: 12/19/2022]
|
14
|
Microperfusion of 3-MPA into the brain augments GABA. Epilepsy Behav 2013; 29:478-84. [PMID: 24094842 PMCID: PMC3939839 DOI: 10.1016/j.yebeh.2013.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/25/2013] [Accepted: 08/26/2013] [Indexed: 11/23/2022]
Abstract
In vivo effects of microperfusion of a GABA synthesis inhibitor (3-MPA) into the striatum and hippocampus on amino acid concentrations and electrical neuronal activity were investigated. Paradoxical elevations in GABA in the striatum (5-fold in anesthetized and 50-fold in awake rats) and hippocampus (2-fold in anesthetized and 15-fold in awake rats) were documented under steady-state concentrations of 3-MPA along with expected increases in glutamate (a 15-fold increase and a 250-fold increase in the striatum of anesthetized and awake rats, respectively; a 7-fold increase and a 25-fold increase in the hippocampus of anesthetized and awake rats, respectively). There was no clear epileptiform or seizure activity. Explanations for the paradoxical increase in GABA are offered, and emphasis is placed on the dependency of disinhibition on the model in which its effects are studied as well as on the prevailing level of activation of the probed network.
Collapse
|
15
|
|
16
|
Abstract
Brain injury during development can have severe, long-term consequences. Using an array of animal models, we have an understanding of the etiology of perinatal brain injury. However, we have only recently begun to address the consequences of endogenous factors such as genetic sex and developmental steroid hormone milieu. Our limited understanding has sometimes led researchers to make over-generalizing and potentially dangerous statements regarding treatment for brain injury. Therefore this review acts as a cautionary tale, speaking to our need to understand the effects of sex and steroid hormone environment on the response to brain trauma in the neonate.
Collapse
Affiliation(s)
- Joseph Nuñez
- Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI 48824, USA.
| |
Collapse
|
17
|
Brooks PL, Peever JH. Identification of the transmitter and receptor mechanisms responsible for REM sleep paralysis. J Neurosci 2012; 32:9785-95. [PMID: 22815493 PMCID: PMC6621291 DOI: 10.1523/jneurosci.0482-12.2012] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 04/25/2012] [Accepted: 05/07/2012] [Indexed: 11/21/2022] Open
Abstract
During REM sleep the CNS is intensely active, but the skeletal motor system is paradoxically forced into a state of muscle paralysis. The mechanisms that trigger REM sleep paralysis are a matter of intense debate. Two competing theories argue that it is caused by either active inhibition or reduced excitation of somatic motoneuron activity. Here, we identify the transmitter and receptor mechanisms that function to silence skeletal muscles during REM sleep. We used behavioral, electrophysiological, receptor pharmacology and neuroanatomical approaches to determine how trigeminal motoneurons and masseter muscles are switched off during REM sleep in rats. We show that a powerful GABA and glycine drive triggers REM paralysis by switching off motoneuron activity. This drive inhibits motoneurons by targeting both metabotropic GABA(B) and ionotropic GABA(A)/glycine receptors. REM paralysis is only reversed when motoneurons are cut off from GABA(B), GABA(A) and glycine receptor-mediated inhibition. Neither metabotropic nor ionotropic receptor mechanisms alone are sufficient for generating REM paralysis. These results demonstrate that multiple receptor mechanisms trigger REM sleep paralysis. Breakdown in normal REM inhibition may underlie common sleep motor pathologies such as REM sleep behavior disorder.
Collapse
Affiliation(s)
- Patricia L Brooks
- Department of Cell and Systems Biology, Systems Neurobiology Laboratory, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | | |
Collapse
|
18
|
Barreto G, White RE, Ouyang Y, Xu L, Giffard RG. Astrocytes: targets for neuroprotection in stroke. Cent Nerv Syst Agents Med Chem 2012; 11:164-73. [PMID: 21521168 DOI: 10.2174/187152411796011303] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/05/2011] [Accepted: 03/09/2011] [Indexed: 02/08/2023]
Abstract
In the past two decades, over 1000 clinical trials have failed to demonstrate a benefit in treating stroke, with the exception of thrombolytics. Although many targets have been pursued, including antioxidants, calcium channel blockers, glutamate receptor blockers, and neurotrophic factors, often the focus has been on neuronal mechanisms of injury. Broader attention to loss and dysfunction of non-neuronal cell types is now required to increase the chance of success. Of the several glial cell types, this review will focus on astrocytes. Astrocytes are the most abundant cell type in the higher mammalian nervous system, and they play key roles in normal CNS physiology and in central nervous system injury and pathology. In the setting of ischemia astrocytes perform multiple functions, some beneficial and some potentially detrimental, making them excellent candidates as therapeutic targets to improve outcome following stroke and in other central nervous system injuries. The older neurocentric view of the central nervous system has changed radically with the growing understanding of the many essential functions of astrocytes. These include K+ buffering, glutamate clearance, brain antioxidant defense, close metabolic coupling with neurons, and modulation of neuronal excitability. In this review, we will focus on those functions of astrocytes that can both protect and endanger neurons, and discuss how manipulating these functions provides a novel and important strategy to enhance neuronal survival and improve outcome following cerebral ischemia.
Collapse
Affiliation(s)
- George Barreto
- Department of Anesthesia, Stanford University School of Medicine, S272, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
19
|
Higuera-Matas A, Miguéns M, Coria SM, Assis MA, Borcel E, del Olmo N, Ambrosio E. Sex-specific disturbances of the glutamate/GABA balance in the hippocampus of adult rats subjected to adolescent cannabinoid exposure. Neuropharmacology 2012; 62:1975-84. [PMID: 22245681 DOI: 10.1016/j.neuropharm.2011.12.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 12/22/2011] [Accepted: 12/23/2011] [Indexed: 10/14/2022]
Abstract
Adolescence is a period of active synaptic remodelling and plasticity and as such, a developmental phase of particular vulnerability to the effects of environmental insults. The endogenous cannabinoid system regulates central nervous system development and cannabinoid exposure during adolescence has been linked to several alterations to hippocampal-dependent processes such as cognition and emotion, which rely on intact glutamatergic and GABAergic systems. Here we show that K(+)-induced γ-amino butyric acid (GABA) release increases in the CA1 hippocampal field of Wistar rats of both sexes that were treated chronically with the cannabinoid agonist CP 55,940 (CP55940) during adolescence. GABA(B) receptors levels also increased in cannabinoid-exposed rats. In addition, CP55940-treated females exhibit reduced GABA transporter gene expression (GAT-1), increased GABA(A) receptor expression, as well as decreased K(+)-induced glutamate release and NMDA receptor levels. CP55940 administration did not affect the glial (EAAT2) or neuronal (EAAT3) glutamate transporter gene expression in either males or females, and nor were any changes in the mGlu5 receptor protein levels observed. Taken together, these results show that while the exacerbated GABA release induced by early cannabinoid exposure may be compensated by an increment in GABA(B) receptors, which normally function as inhibitory autoreceptors, adolescent cannabinoid exposure in the females disturbs the normal balance between glutamate and GABA transmission. These observations may provide important insight into the neuronal basis of the well-documented alterations in cognitive and emotional processes induced by adolescent cannabinoid exposure.
Collapse
Affiliation(s)
- Alejandro Higuera-Matas
- Department of Psychobiology, School of Psychology, UNED, C/Juan del Rosal no 10, 28040 Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
20
|
Zheng M, Kang YM, Liu W, Zang WJ, Bao CY, Qin DN. Inhibition of cyclooxygenase-2 reduces hypothalamic excitation in rats with adriamycin-induced heart failure. PLoS One 2012; 7:e48771. [PMID: 23152801 PMCID: PMC3496718 DOI: 10.1371/journal.pone.0048771] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/04/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The paraventricular nucleus (PVN) of the hypothalamus plays an important role in the progression of heart failure (HF). We investigated whether cyclooxygenase-2 (COX-2) inhibition in the PVN attenuates the activities of sympathetic nervous system (SNS) and renin-angiotensin system (RAS) in rats with adriamycin-induced heart failure. METHODOLOGY/PRINCIPAL FINDING Heart failure was induced by intraperitoneal injection of adriamycin over a period of 2 weeks (cumulative dose of 15 mg/kg). On day 19, rats received intragastric administration daily with either COX-2 inhibitor celecoxib (CLB) or normal saline. Treatment with CLB reduced mortality and attenuated both myocardial atrophy and pulmonary congestion in HF rats. Compared with the HF rats, ventricle to body weight (VW/BW) and lung to body weight (LW/BW) ratios, heart rate (HR), left ventricular end-diastolic pressure (LVEDP), left ventricular peak systolic pressure (LVPSP) and maximum rate of change in left ventricular pressure (LV±dp/dtmax) were improved in HF+CLB rats. Angiotensin II (ANG II), norepinephrine (NE), COX-2 and glutamate (Glu) in the PVN were increased in HF rats. HF rats had higher levels of ANG II and NE in plasma, higher level of ANG II in myocardium, and lower levels of ANP in plasma and myocardium. Treatment with CLB attenuated these HF-induced changes. HF rats had more COX-2-positive neurons and more corticotropin releasing hormone (CRH) positive neurons in the PVN than did control rats. Treatment with CLB decreased COX-2-positive neurons and CRH positive neurons in the PVN of HF rats. CONCLUSIONS These results suggest that PVN COX-2 may be an intermediary step for PVN neuronal activation and excitatory neurotransmitter release, which further contributes to sympathoexcitation and RAS activation in adriamycin-induced heart failure. Treatment with COX-2 inhibitor attenuates sympathoexcitation and RAS activation in adriamycin-induced heart failure.
Collapse
Affiliation(s)
- Min Zheng
- Department of Physiology, Shantou University Medical College, Shantou, P. R. China
- Department of Biomedical Engineering, Hubei University of Science and Techonology, Xianning, P. R. China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Medicine, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, P. R. China
- * E-mail: (DNQ); (YMK)
| | - Wei Liu
- Department of Physiology, Shantou University Medical College, Shantou, P. R. China
| | - Wei-Jin Zang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi’an, P. R. China
| | - Cui-Yu Bao
- Department of Biomedical Engineering, Hubei University of Science and Techonology, Xianning, P. R. China
| | - Da-Nian Qin
- Department of Physiology, Shantou University Medical College, Shantou, P. R. China
- * E-mail: (DNQ); (YMK)
| |
Collapse
|
21
|
Chen Y, Zhao B, Huang X, Zhan J, Zhao Y, Zhou M, Guo L. Purification and neuroprotective effects of polysaccharides from Opuntia Milpa Alta in cultured cortical neurons. Int J Biol Macromol 2011; 49:681-7. [PMID: 21763720 DOI: 10.1016/j.ijbiomac.2011.06.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 06/24/2011] [Accepted: 06/30/2011] [Indexed: 10/18/2022]
Abstract
Opuntia is a traditional plant from China with medicinal applications. In this experiment, polysaccharides from Opuntia Milpa Alta (MAPs) were analyzed using gas chromatograph-mass spectrometer (GC-MS) method and result showed that MAPs contained mannose (6.37%), rhamnose (14.94%), xylose (1.99%), arabinose (24.07%), galactose (38.25%), ribose (2.63%) and glucose (11.48%). The neuroprotective effects of MAPs were evaluated at the mechanistic level in vitro models of cerebral ischemic injury. In vitro oxygen/glucose deprivation (OGD) model, MAPs (0.5 μg/ml, 5 μg/ml, 50 μg/ml) effectively increased cell viability by methyl thiazolyl tetrazolium (MTT) assay, inhibited cell cytotoxicity by lactate dehydrogenase (LDH) assay, reduced neuronal cell death, suppressed the production of intracellular reactive oxygen species (ROS), decreased of intracellular free Ca(2+) concentrations ([Ca(2+)](i)), and reduced extracellular glutamate level. Therefore, MAPs might prevent intracellular calcium overload and decreased glutamate excitotoxicity, both of which can cause neuronal injury and death in vitro models of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yang Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | | | | | | | | | | | | |
Collapse
|
22
|
Barreto GE, Gonzalez J, Torres Y, Morales L. Astrocytic-neuronal crosstalk: implications for neuroprotection from brain injury. Neurosci Res 2011; 71:107-13. [PMID: 21693140 DOI: 10.1016/j.neures.2011.06.004] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 04/28/2011] [Accepted: 05/16/2011] [Indexed: 10/18/2022]
Abstract
The older neurocentric view of the central nervous system (CNS) has changed radically with the growing understanding of the many essential functions of astrocytes. Advances in our understanding of astrocytes include new observations about their structure, organization, function and supportive actions to other cells. Although the contribution of astrocytes to the process of brain injury has not been clearly defined, it is thought that their ability to provide support to neurons after cerebral damage is critical. Astrocytes play a fundamental role in the pathogenesis of brain injury-associated neuronal death, and this secondary injury is primarily a consequence of the failure of astrocytes to support the essential metabolic needs of neurons. These needs include K+ buffering, glutamate clearance, brain antioxidant defense, close metabolic coupling with neurons, and the modulation of neuronal excitability. In this review, we will focus on astrocytic activities that can both protect and endanger neurons, and discuss how manipulating these functions provides a novel and important strategy to enhance neuronal survival and improve the outcome following brain injury.
Collapse
Affiliation(s)
- George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia.
| | | | | | | |
Collapse
|
23
|
Chi OZ, Hunter C, Liu X, Chi Y, Weiss HR. Effects of GABA(A) receptor blockade on regional cerebral blood flow and blood-brain barrier disruption in focal cerebral ischemia. J Neurol Sci 2010; 301:66-70. [PMID: 21094956 DOI: 10.1016/j.jns.2010.10.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/14/2010] [Accepted: 10/22/2010] [Indexed: 11/18/2022]
Abstract
In cerebral ischemia, transmission by the inhibitory neurotransmitter, γ-aminobutyric acid (GABA) is altered. This study was performed to determine whether blockade of GABA(A) receptor would affect regional cerebral blood flow (rCBF) and blood-brain barrier (BBB) permeability in a focal ischemic area of the brain. Rats were anesthetized with isoflurane and mechanically ventilated. Fifteen minutes after a permanent middle cerebral artery (MCA) occlusion, one half of the rats were infused with bicuculline 1mg/kg/min iv for 2 min followed by 0.1mg/kg/min iv to the end of the experiment. The other half were infused with normal saline. At one hour after MCA occlusion, rCBF was determined using ¹⁴C-iodoantipyrine and BBB permeability was determined by measuring the transfer coefficient (Ki) of ¹⁴C-α-aminoisobutyric acid. With MCA occlusion, rCBF was decreased in the ischemic cortex (IC) (-70%) in the control rats. In the bicuculline treated rats, the rCBF of the IC was lower (-48%) than the contralateral cortex but higher than the rCBF of the IC of the control rats (+55%). MCA occlusion increased Ki in the IC of the control rats (+72%) and bicuculline administration increased Ki further (+53%) in the IC. Blockade of GABA(A) receptors did not significantly affect rCBF or BBB permeability in the non-ischemic brain regions under isoflurane anesthesia. Our data demonstrated that blockade of GABA(A) receptors increased rCBF and enhanced the BBB disruption in focal cerebral ischemia. Our data suggest that GABA(A) receptors are involved, at least in part, in modulating rCBF and BBB disruption in focal cerebral ischemia.
Collapse
Affiliation(s)
- Oak Z Chi
- Department of Anesthesia, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901-1977, USA.
| | | | | | | | | |
Collapse
|
24
|
Zhang JH, Fung SJ, Xi M, Sampogna S, Chase MH. Prevention of apnea-induced apoptosis in NREM- and REM-generating nuclei of adult guinea pigs. Brain Res 2010; 1347:161-9. [PMID: 20515665 DOI: 10.1016/j.brainres.2010.05.078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 05/23/2010] [Accepted: 05/25/2010] [Indexed: 12/31/2022]
Abstract
The present study was designed to investigate the effects of recurrent periods of apnea/hypoxia on the morphology of neurons in sites that control NREM and REM sleep. In addition, we determined whether the administration of a GABA agonist, eszopiclone, was capable of preventing the degenerative, i.e., apoptotic, sequelae of hypoxia in these sleep-promoting neurons. Adult guinea pigs were divided into control (normoxic) and hypoxic groups; a separate group of hypoxic animals was administered eszopiclone. Recurrent periods of hypoxia and normoxia lasted for a duration of 3h. Subsequently, the brains were sectioned, and areas in the CNS that control NREM sleep as well as REM sleep were examined after staining with an antibody raised against single-stranded DNA, which labels apoptotic neurons. In the group of control (normoxic) animals, apoptotic neurons were not observed in CNS regions that control NREM or REM sleep. In hypoxic animals, a large number of apoptotic neurons were found in the preceding regions. In the hypoxic animals that were administered eszopiclone, there were almost no apoptotic neurons in the brain regions that control NREM or REM sleep. These results demonstrate that recurrent periods of apnea induce extensive apoptosis in CNS nuclei that control NREM and REM sleep and that eszopiclone is capable of preventing neuronal degeneration in these sites. We suggest that the degeneration of neurons in sites that control the states of sleep is responsible for those sleep disturbances that arise as a consequence of hypoxia in individuals with sleep-related breathing disorders.
Collapse
Affiliation(s)
- Jian-Hua Zhang
- WebSciences International, 1251 Westwood Blvd., Los Angeles, CA 90024, USA.
| | | | | | | | | |
Collapse
|
25
|
Cheng CY, Su SY, Tang NY, Ho TY, Lo WY, Hsieh CL. Ferulic acid inhibits nitric oxide-induced apoptosis by enhancing GABA(B1) receptor expression in transient focal cerebral ischemia in rats. Acta Pharmacol Sin 2010; 31:889-99. [PMID: 20644551 PMCID: PMC4007809 DOI: 10.1038/aps.2010.66] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 05/06/2010] [Indexed: 12/20/2022]
Abstract
AIM Ferulic acid (4-hydroxy-3-methoxycinnamic acid, FA) provides neuroprotection against apoptosis in a transient middle cerebral artery occlusion (MCAo) model. This study was to further investigate the anti-apoptotic effect of FA during reperfusion after cerebral ischemia. METHODS Rats were subjected to 90 min of cerebral ischemia followed by 3 or 24 h of reperfusion after which they were sacrificed. RESULTS Intravenous FA (100 mg/kg) administered immediately after middle cerebral artery occlusion (MCAo) or 2 h after reperfusion effectively abrogated the elevation of postsynaptic density-95 (PSD-95), neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase (iNOS), nitrotyrosine, and cleaved caspase-3 levels as well as apoptosis in the ischemic cortex at 24 h of reperfusion. FA further inhibited Bax translocation, cytochrome c release, and p38 mitogen-activated protein (MAP) kinase phosphorylation. Moreover, FA enhanced the expression of gamma-aminobutyric acid type B receptor subunit 1 (GABA(B1)) in the ischemic cortex at 3 and 24 h of reperfusion. In addition, nitrotyrosine-positive cells colocalized with cleaved caspase-3-positive cells, and phospho-p38 MAP kinase-positive cells colocalized with nitrotyrosine- and Bax-positive cells, indicating a positive relationship among the expression of nitrotyrosine, phospho-p38 MAP kinase, Bax, and cleaved caspase-3. The mutually exclusive expression of GABA(B1) and nitrotyrosine revealed that there is a negative correlation between GABA(B1) and nitrotyrosine expression profiles. Additionally, pretreatment with saclofen, a GABA(B) receptor antagonist, abolished the neuroprotection of FA against nitric oxide (NO)-induced apoptosis. CONCLUSION FA significantly enhances GABA(B1) receptor expression at early reperfusion and thereby provides neuroprotection against p38 MAP kinase-mediated NO-induced apoptosis at 24 h of reperfusion.
Collapse
Affiliation(s)
- Chin-yi Cheng
- Graduate Institute of Acupuncture Science, China Medical University 40402, Taichung, Taiwan, China
- Acupuncture Research Center, China Medical University 40402, Taichung, Taiwan, China
| | - Shan-yu Su
- School of Chinese Medicine, China Medical University 40402, Taichung, Taiwan, China
- Department of Chinese Medicine, China Medical University Hospital 40447, Taichung, Taiwan, China
| | - Nou-ying Tang
- School of Chinese Medicine, China Medical University 40402, Taichung, Taiwan, China
| | - Tin-yun Ho
- School of Chinese Medicine, China Medical University 40402, Taichung, Taiwan, China
| | - Wan-yu Lo
- Graduate Institute of Integrated Medicine, China Medical University 40402, Taichung, Taiwan, China
| | - Ching-liang Hsieh
- Graduate Institute of Acupuncture Science, China Medical University 40402, Taichung, Taiwan, China
- Acupuncture Research Center, China Medical University 40402, Taichung, Taiwan, China
- Department of Chinese Medicine, China Medical University Hospital 40447, Taichung, Taiwan, China
| |
Collapse
|
26
|
Mathew J, Soman S, Sadanandan J, Paulose CS. Decreased GABA receptor in the striatum and spatial recognition memory deficit in epileptic rats: effect of Bacopa monnieri and bacoside-A. JOURNAL OF ETHNOPHARMACOLOGY 2010; 130:255-261. [PMID: 20451596 DOI: 10.1016/j.jep.2010.04.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 04/10/2010] [Accepted: 04/28/2010] [Indexed: 05/29/2023]
Abstract
AIM OF THE STUDY Gamma-aminobutyric acid A receptors are the principal mediators of synaptic inhibition in striatal neurons and play an important role in preventing the spreading of seizures through the striatum. In the present study, effect of Bacopa monnieri (L.) Pennel and its active component bacoside-A on spatial recognition memory deficit and alterations of GABA receptor in the striatum of epileptic rats were investigated. MATERIALS AND METHODS Total GABA and GABA(A) receptor numbers in the control and epileptic rats were evaluated using [(3)H]GABA and [(3)H]bicuculline binding. GABA(Aalpha1,) GABA(Aalpha5,) GABA(Agamma3) and GABA(Adelta) gene expressions were studied. Behavioral performance was assed using Y-maze. RESULTS Scatchard analysis of [(3)H]GABA and [(3)H]bicuculline in the striatum of epileptic rats showed significant decrease in B(max) compared to control. Real-Time PCR amplification of GABA(A) receptor subunits such as GABA(Aalpha1,) GABA(Aalpha5) and GABA(Adelta), were down regulated (p<0.001) in the striatum of epileptic rats compared to control. Epileptic rats have deficit in Y-maze performance. Bacopa monnieri and bacoside-A treatment reversed these changes to near control. CONCLUSION Our results suggest that decreased GABA receptors in the striatum have an important role in epilepsy associated motor learning deficits and Bacopa monnieri and bacoside-A has a beneficial effect in the management of epilepsy.
Collapse
Affiliation(s)
- Jobin Mathew
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Cochin, Kerala, India
| | | | | | | |
Collapse
|
27
|
McLamore ES, Mohanty S, Shi J, Claussen J, Jedlicka SS, Rickus JL, Porterfield DM. A self-referencing glutamate biosensor for measuring real time neuronal glutamate flux. J Neurosci Methods 2010; 189:14-22. [PMID: 20298719 DOI: 10.1016/j.jneumeth.2010.03.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/14/2010] [Accepted: 03/02/2010] [Indexed: 01/21/2023]
Abstract
Quantification of neurotransmitter transport dynamics is hindered by a lack of sufficient tools to directly monitor bioactive flux under physiological conditions. Traditional techniques for studying neurotransmitter release/uptake require inferences from non-selective electrical recordings, are invasive/destructive, and/or suffer from poor temporal resolution. Recent advances in electrochemical biosensors have enhanced in vitro and in vivo detection of neurotransmitter concentration under physiological/pathophysiological conditions. The use of enzymatic biosensors with performance enhancing materials (e.g., carbon nanotubes) has been a major focus for many of these advances. However, these techniques are not used as mainstream neuroscience research tools, due to relatively low sensitivity, excessive drift/noise, low signal-to-noise ratio, and inability to quantify rapid neurochemical kinetics during synaptic transmission. A sensing technique known as self-referencing overcomes many of these problems, and allows non-invasive quantification of biophysical transport. This work presents a self-referencing CNT modified glutamate oxidase biosensor for monitoring glutamate flux near neural/neuronal cells. Concentration of basal glutamate was similar to other in vivo and in vitro measurements. The biosensor was used in self-referencing (oscillating) mode to measure net glutamate flux near neural cells during electrical stimulation. Prior to stimulation, the average influx was 33.9+/-6.4 fmol cm(-2)s(-1)). Glutamate efflux took place immediately following stimulation, and was always followed by uptake in the 50-150 fmol cm(-2)s(-1) range. Uptake was inhibited using threo-beta-benzyloxyaspartate, and average surface flux in replicate cells (1.1+/-7.4 fmol cm(-2)s(-1)) was significantly lower than uninhibited cells. The technique is extremely valuable for studying neuropathological conditions related to neurotransmission under dynamic physiological conditions.
Collapse
Affiliation(s)
- E S McLamore
- Department of Agricultural & Biological Engineering, Purdue University, West Lafayette, IN 47906, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Rotge JY, Aouizerate B, Tignol J, Bioulac B, Burbaud P, Guehl D. The glutamate-based genetic immune hypothesis in obsessive-compulsive disorder. An integrative approach from genes to symptoms. Neuroscience 2010; 165:408-17. [PMID: 19861150 DOI: 10.1016/j.neuroscience.2009.10.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 10/21/2009] [Accepted: 10/21/2009] [Indexed: 10/20/2022]
Abstract
Recent advances in multiple areas of research have contributed to the identification of several pathophysiological factors underlying obsessive-compulsive disorder (OCD). In particular, the glutamate transporter gene SLC1A1 has been associated with the diagnosis of OCD. Immunological and infectious studies have reported alterations of the immune system and the presence of immune complexes directed against the Borna disease virus in OCD patients. In addition, neuroimaging of OCD patients has demonstrated abnormalities in the anterior cingulate cortex, orbitofrontal cortex, thalamus, and the basal ganglia. Neuropsychological assessments have found several cognitive disruptions that have been identified in OCD, especially impairments in cognitive flexibility. Here, we attempt to bridge the gap between these remarkable findings through several previously unpredicted pathophysiological mechanisms. We propose an integrative hypothesis that indicates how genetic and environmental factors may contribute to the structural and functional alterations of cortico-subcortical circuits, leading to the characteristic cognitive disruptions underlying OCD symptoms.
Collapse
Affiliation(s)
- J Y Rotge
- Laboratoire Mouvement Adaptation Cognition, CNRS UMR 5227, Université Bordeaux 2, Bordeaux, France.
| | | | | | | | | | | |
Collapse
|
29
|
Pu F, Kaneko T, Enoki M, Irie K, Okamoto T, Sei Y, Egashira N, Oishi R, Mishima K, Kamimura H, Iwasaki K, Fujiwara M. Ameliorating effects of Kangen-karyu on neuronal damage in rats subjected to repeated cerebral ischemia. J Nat Med 2010; 64:167-74. [DOI: 10.1007/s11418-010-0392-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 01/07/2010] [Indexed: 01/10/2023]
|
30
|
Sander S, Richter F, Raymond R, Diwan M, Lange N, Nobrega J, Richter A. Pharmacological and autoradiographic studies on the pathophysiological role of GABAB receptors in the dystonic hamster: pronounced antidystonic effects of baclofen after striatal injections. Neuroscience 2009; 162:423-30. [DOI: 10.1016/j.neuroscience.2009.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 05/04/2009] [Accepted: 05/04/2009] [Indexed: 12/18/2022]
|
31
|
Cimarosti H, Kantamneni S, Henley JM. Ischaemia differentially regulates GABA(B) receptor subunits in organotypic hippocampal slice cultures. Neuropharmacology 2009; 56:1088-96. [PMID: 19328818 PMCID: PMC3309028 DOI: 10.1016/j.neuropharm.2009.03.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 02/19/2009] [Accepted: 03/18/2009] [Indexed: 02/04/2023]
Abstract
Reduced synaptic inhibition due to dysfunction of ionotropic GABA(A) receptors has been proposed as one factor in cerebral ischaemia-induced excitotoxic cell death. However, the participation of the inhibitory metabotropic GABA(B) receptors in these pathological processes has not been extensively investigated. We used oxygen-glucose deprivation (OGD) and NMDA-induced excitotoxicity as models to investigate whether ischaemia-like challenges alter the protein levels of GABA(B1) and GABA(B2) receptor subunits in rat organotypic hippocampal slice cultures. Twenty-four hours after the insult both OGD and NMDA produced a marked decrease in the total levels of GABA(B2) (approximately 75%), while there was no significant change in the levels of GABA(B1) after OGD, but an increase after NMDA treatment (approximately 100%). The GABA(B) receptor agonist baclofen (100 microM) was neuroprotective following OGD or NMDA treatment if added before or during the insult. GABA(B) receptors comprise heterodimers of GABA(B1) and GABA(B2) subunits and our results suggest that the separate subunits are independently regulated in response to extreme neuronal stress. However, because GABA(B2) is required for functional surface expression, down-regulation of this subunit removes an important inhibitory feedback mechanism under pathological conditions.
Collapse
Affiliation(s)
| | | | - Jeremy M. Henley
- Department of Anatomy, MRC Centre for Synaptic Plasticity, School of Medical Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
32
|
Pre-ischemic treadmill training affects glutamate and gamma aminobutyric acid levels in the striatal dialysate of a rat model of cerebral ischemia. Life Sci 2009; 84:505-11. [DOI: 10.1016/j.lfs.2009.01.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 12/05/2008] [Accepted: 01/26/2009] [Indexed: 11/17/2022]
|
33
|
He Z, Lu Q, Xu X, Huang L, Chen J, Guo L. DDPH ameliorated oxygen and glucose deprivation-induced injury in rat hippocampal neurons via interrupting Ca2+ overload and glutamate release. Eur J Pharmacol 2008; 603:50-5. [PMID: 19105952 DOI: 10.1016/j.ejphar.2008.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 11/19/2008] [Accepted: 12/02/2008] [Indexed: 11/26/2022]
Abstract
Our previous work has demonstrated that DDPH (1-(2, 6-dimethylphenoxy)-2-(3, 4-dimethoxyphenylethylamino) propane hydrochloride), a competitive alpha(1)-adrenoceptor antagonist, could improve cognitive deficits, reduce histopathological damage and facilitate synaptic plasticity in vivo possibly via increasing NR2B (NMDA receptor 2B) expression and antioxidation of DDPH itself. The present study further evaluated effects of DDPH on OGD (Oxygen and glucose deprivation)-induced neuronal damage in rat primary hippocampal cells. The addition of DDPH to the cultured cells 12 h before OGD for 4 h significantly reduced neuronal damage as determined by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and LDH (lactate dehydrogenase) release experiments. The effects of DDPH on intracellular calcium concentration were explored by Fura-2 based calcium imaging techniques and results showed that DDPH at the dosages of 5 microM and 10 microM suppressed the increase of intracellular calcium ([Ca(2+)](i)) stimulated by 50 mM KCl in Ca(2+)-containing extracellular solutions. However, DDPH couldn't suppress the increase of [Ca(2+)](i) induced by both 50 microM glutamate in Ca(2+)-containing extracellular solutions and 20 microM ATP (Adenosine Triphosphate) in Ca(2+)-free solution. These results indicated that DDPH prevented [Ca(2+)](i) overload in hippocampal neurons by blocking Ca(2+) influx (voltage-dependent calcium channel) but not Ca(2+) mobilization from the intracellular Ca(2+) store in endoplasm reticulum (ER). We also demonstrated that DDPH could decrease glutamate release when hippocampal cells were subjected to OGD. These observations demonstrated that DDPH protected hippocampal neurons against OGD-induced damage by preventing the Ca(2+) influx and decreasing glutamate release.
Collapse
Affiliation(s)
- Zhi He
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | | | | | | | | | | |
Collapse
|
34
|
Inhibition of intracellular Ca2+ release by a Rho-kinase inhibitor for the treatment of ischemic damage in primary cultured rat hippocampal neurons. Eur J Pharmacol 2008; 602:238-44. [PMID: 19070614 DOI: 10.1016/j.ejphar.2008.11.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 11/06/2008] [Accepted: 11/24/2008] [Indexed: 11/21/2022]
Abstract
The effects of hydroxy fasudil, a specific Rho-kinase inhibitor, on behavior and brain neuronal activity in animal studies have been described previously. However, whether a Rho-kinase inhibitor can directly protect neurons against ischemic damage and the molecular mechanisms underlying these effects are poorly understood. The present work was designed to investigate the effect of hydroxy fasudil against oxygen-glucose deprivation (OGD) induced acute neuronal injury and the underlying mechanisms in vitro. Pretreatment with hydroxy fasudil at 5 and 10 microM could concentration-dependently improve cell viability and decrease Lactate dehydrogenase (LDH) level in extracellular solution of neurons suffered from OGD either in Ca(2+)-containing or Ca(2+)-free culture medium. Moreover, we found that abnormal elevation of extracellular glutamate (Glu) level induced by OGD was markedly repressed by hydroxy fasudil as measured by high performance liquid chromatography (HPLC). Using Fura-2 based calcium imaging techniques, we further demonstrated that preincubation with hydroxy fasudil suppressed the increase of [Ca(2+)](i) induced by 50 microM Glu and 20 microM ATP, but had no effect on the increase of [Ca(2+)](i) induced by 50 mM KCl. These data demonstrated that the neuroprotective effect of hydroxy fasudil was attributed to repressing Glu excitotoxicity and ischemic induced calcium overload by inhibiting Ca(2+) release from Ca(2+) stores rather than by inhibiting Ca(2+) influx via receptor-operated or voltage-dependent calcium channel.
Collapse
|
35
|
Temporal patterns of motor behavioural improvements by MK-801 in Mongolian gerbils submitted to different duration of global cerebral ischemia. Behav Brain Res 2008; 194:72-8. [DOI: 10.1016/j.bbr.2008.06.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/17/2008] [Accepted: 06/20/2008] [Indexed: 01/09/2023]
|
36
|
Neal-Perry GS, Zeevalk GD, Shu J, Etgen AM. Restoration of the luteinizing hormone surge in middle-aged female rats by altering the balance of GABA and glutamate transmission in the medial preoptic area. Biol Reprod 2008; 79:878-88. [PMID: 18667749 DOI: 10.1095/biolreprod.108.069831] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Hypothalamic glutamate and gamma-aminobutyric acid (GABA) neurotransmission are involved in the ovarian hormone-induced GnRH-LH surge in rodents. We previously reported that middle-aged rats have significantly less glutamate release in the medial preoptic area than young rats on the day of the LH surge. The present study tested the hypothesis that the delayed and attenuated LH surge in ovariohysterectomized middle-aged rats primed with ovarian steroids results from reduced hypothalamic glutamate and increased GABA(A) neurotransmission. Microdialysis results show that middle-aged rats with attenuated LH surges had reduced extracellular glutamate and increased extracellular GABA levels in the medial preoptic area compared with young rats. Blocking GABA(A) receptors with bicuculline or inhibiting synaptic glutamate reuptake with L-trans-pyrrolidine-2,4-dicarboxylic acid increased extracellular Glu in the medial preoptic area and partially restored LH surge amplitude in middle-aged rats without altering LH surge onset. Complete recovery of LH surge amplitude was observed in middle-aged rats treated with the combination of bicuculline and L-trans-pyrrolidine-2,4-dicarboxylic acid. This treatment also restored the extracellular glutamate:GABA ratio in the medial preoptic area of middle-aged rats to the level of young rats. Immunoblot analysis revealed that estradiol and progesterone treatment reduced SLC32A1(formerly known as vesicular GABA transporter) levels and increased SLC17A6 (formerly known as vesicular glutamate transporter 2) levels in the anterior hypothalamus of ovariohysterectomized young but not middle-aged rats. These data suggest that both reduced availability of glutamate and increased activation of GABA(A) receptors under estrogen-positive feedback conditions contribute to the age-related delay in onset and attenuated amplitude of the LH surge.
Collapse
Affiliation(s)
- Genevieve S Neal-Perry
- Department of Obstetrics and Gynecology, Division of Reproductive Medicine and Infertility, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | | | |
Collapse
|
37
|
Málly J, Dinya E. Recovery of motor disability and spasticity in post-stroke after repetitive transcranial magnetic stimulation (rTMS). Brain Res Bull 2008; 76:388-95. [DOI: 10.1016/j.brainresbull.2007.11.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 11/28/2007] [Accepted: 11/28/2007] [Indexed: 12/21/2022]
|
38
|
Hertz L. Bioenergetics of cerebral ischemia: a cellular perspective. Neuropharmacology 2008; 55:289-309. [PMID: 18639906 DOI: 10.1016/j.neuropharm.2008.05.023] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 05/14/2008] [Accepted: 05/14/2008] [Indexed: 12/27/2022]
Abstract
In cerebral ischemia survival of neurons, astrocytes, oligodendrocytes and endothelial cells is threatened during energy deprivation and/or following re-supply of oxygen and glucose. After a brief summary of characteristics of different cells types, emphasizing the dependence of all on oxidative metabolism, the bioenergetics of focal and global ischemia is discussed, distinguishing between events during energy deprivation and subsequent recovery attempt after re-circulation. Gray and white matter ischemia are described separately, and distinctions are made between mature and immature brains. Next comes a description of bioenergetics in individual cell types in culture during oxygen/glucose deprivation or exposure to metabolic inhibitors and following re-establishment of normal aerated conditions. Due to their expression of NMDA and non-NMDA receptors neurons and oligodendrocytes are exquisitely sensitive to excitotoxicity by glutamate, which reaches high extracellular concentrations in ischemic brain for several reasons, including failing astrocytic uptake. Excitotoxicity kills brain cells by energetic exhaustion (due to Na(+) extrusion after channel-mediated entry) combined with mitochondrial Ca(2+)-mediated injury and formation of reactive oxygen species. Many (but not all) astrocytes survive energy deprivation for extended periods, but after return to aerated conditions they are vulnerable to mitochondrial damage by cytoplasmic/mitochondrial Ca(2+) overload and to NAD(+) deficiency. Ca(2+) overload is established by reversal of Na(+)/Ca(2+) exchangers following Na(+) accumulation during Na(+)-K(+)-Cl(-) cotransporter stimulation or pH regulation, compensating for excessive acid production. NAD(+) deficiency inhibits glycolysis and eventually oxidative metabolism, secondary to poly(ADP-ribose)polymerase (PARP) activity following DNA damage. Hyperglycemia can be beneficial for neurons but increases astrocytic death due to enhanced acidosis.
Collapse
Affiliation(s)
- Leif Hertz
- College of Basic Medical Sciences, China Medical University, Shenyang, PR China.
| |
Collapse
|
39
|
Salazar M, Pariente JA, Salido GM, González A. Ethanol induces glutamate secretion by Ca2+ mobilization and ROS generation in rat hippocampal astrocytes. Neurochem Int 2008; 52:1061-1067. [PMID: 18082912 DOI: 10.1016/j.neuint.2007.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 10/30/2007] [Accepted: 11/07/2007] [Indexed: 12/23/2022]
Abstract
In this study we have investigated the effect of ethanol on [Ca2+]c by microfluorimetry and glutamate secretion using an enzyme-linked system, in rat hippocampal astrocytes in culture. Our results show that ethanol (1-200 mM) evoked a dose-dependent increase in glutamate secretion. 50 mM ethanol, a concentration within the range of blood alcohol levels in intoxicated humans, induced a release of Ca2+ from intracellular stores in the form of oscillations. Ca2+-mobilizing effect of ethanol was not prevented by preincubation of cells in the presence of 2 mM of the antioxidant dithiothreitol. Ethanol-evoked glutamate secretion was reduced when extracellular Ca2+ was omitted (medium containing 0.5 mM EGTA) and following preincubation of astrocytes in the presence of the intracellular Ca2+ chelator 1,2-bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid tetraacetoxy-methyl ester (10 microM). Preincubation of astrocytes in the presence of 2 mM of the antioxidant dithiothreitol significantly reduced ethanol-evoked glutamate secretion. Finally, preincubation of astrocytes in the presence of bafilomycin (50 nM) significantly reduced ethanol-induced neurotransmitter release, indicating that exocytosis is involved in glutamate secretion. In conclusion, our results suggest that ethanol mobilizes Ca2+ from intracellular stores, and stimulates a Ca2+-dependent glutamate secretion, probably involving reactive oxygen species production, and therefore creating a situation potentially leading to neurotoxicity in the hippocampus.
Collapse
Affiliation(s)
- Miguel Salazar
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Avenida Universidad s/n, E-10071, Cáceres, Spain
| | | | | | | |
Collapse
|
40
|
Darbin O, Wichmann T. Effects of Striatal GABAA-Receptor Blockade on Striatal and Cortical Activity in Monkeys. J Neurophysiol 2008; 99:1294-305. [DOI: 10.1152/jn.01191.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To elucidate the role of ambient striatal γ-aminobutyric acid (GABA) in the regulation of neuronal activity in the basal ganglia–thalamocortical circuits, we studied the effects of blocking striatal GABAA receptors on the electrical activities of single striatal neurons, on local field potentials (LFPs) in the striatum, and on motor cortical electroencephalograms (EEGs) in two monkeys. Striatal LFPs were recorded with a device that allowed us to simultaneously record field potentials and apply drugs by reverse microdialysis at the same site. Administration of the GABAA-receptor antagonist gabazine (SR95531, 10 and 500 μM) induced large-amplitude LFP fluctuations at the infusion site, occurring every 2–5 s for about 2 h after the start of the 20-min drug administration. These events were prevented by cotreatment with a GABAA-receptor agonist (muscimol, 100 μM) or a combination of ionotropic glutamate receptor antagonists (CNQX and MK-801, each given at 100 μM). Gabazine (10 μM) also increased the firing of single neurons recorded close to the injection site, but in most cases there was no correlation between single-neuron activity and the concomitantly recorded LFP signals from the same striatal region. In contrast, intrastriatal application of gabazine increased the correlation between striatal LFPs and EEG, and resulted in the appearance of recurrent EEG events that were temporally related to the striatal LFP events. These data provide evidence that a GABAergic “tone” in the monkey striatum controls the spontaneous activity of striatal neurons, as well as the level of striatal and cortical synchrony.
Collapse
|