1
|
Nascimento AA, Pereira-Figueiredo D, Borges-Martins VP, Kubrusly RC, Calaza KC. GABAergic system and chloride cotransporters as potential therapeutic targets to mitigate cell death in ischemia. J Neurosci Res 2024; 102:e25355. [PMID: 38808645 DOI: 10.1002/jnr.25355] [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: 08/25/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Gamma aminobutyric acid (GABA) is a critical inhibitory neurotransmitter in the central nervous system that plays a vital role in modulating neuronal excitability. Dysregulation of GABAergic signaling, particularly involving the cotransporters NKCC1 and KCC2, has been implicated in various pathologies, including epilepsy, schizophrenia, autism spectrum disorder, Down syndrome, and ischemia. NKCC1 facilitates chloride influx, whereas KCC2 mediates chloride efflux via potassium gradient. Altered expression and function of these cotransporters have been associated with excitotoxicity, inflammation, and cellular death in ischemic events characterized by reduced cerebral blood flow, leading to compromised tissue metabolism and subsequent cell death. NKCC1 inhibition has emerged as a potential therapeutic approach to attenuate intracellular chloride accumulation and mitigate neuronal damage during ischemic events. Similarly, targeting KCC2, which regulates chloride efflux, holds promise for improving outcomes and reducing neuronal damage under ischemic conditions. This review emphasizes the critical roles of GABA, NKCC1, and KCC2 in ischemic pathologies and their potential as therapeutic targets. Inhibiting or modulating the activity of these cotransporters represents a promising strategy for reducing neuronal damage, preventing excitotoxicity, and improving neurological outcomes following ischemic events. Furthermore, exploring the interactions between natural compounds and NKCC1/KCC2 provides additional avenues for potential therapeutic interventions for ischemic injury.
Collapse
Affiliation(s)
- A A Nascimento
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - D Pereira-Figueiredo
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| | - V P Borges-Martins
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - R C Kubrusly
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
2
|
Garcia JD, Wolfe SE, Stewart AR, Tiemeier E, Gookin SE, Guerrero MB, Quillinan N, Smith KR. Distinct mechanisms drive sequential internalization and degradation of GABA ARs during global ischemia and reperfusion injury. iScience 2023; 26:108061. [PMID: 37860758 PMCID: PMC10582478 DOI: 10.1016/j.isci.2023.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/30/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Synaptic inhibition is critical for controlling neuronal excitability and function. During global cerebral ischemia (GCI), inhibitory synapses are rapidly eliminated, causing hyper-excitability which contributes to cell-death and the pathophysiology of disease. Sequential disassembly of inhibitory synapses begins within minutes of ischemia onset: GABAARs are rapidly trafficked away from the synapse, the gephyrin scaffold is removed, followed by loss of the presynaptic terminal. GABAARs are endocytosed during GCI, but how this process accompanies synapse disassembly remains unclear. Here, we define the precise trafficking itinerary of GABAARs during the initial stages of GCI, placing them in the context of rapid synapse elimination. Ischemia-induced GABAAR internalization quickly follows their initial dispersal from the synapse, and is controlled by PP1α signaling. During reperfusion injury, GABAARs are then trafficked to lysosomes for degradation, leading to permanent removal of synaptic GABAARs and contributing to the profound reduction in synaptic inhibition observed hours following ischemia onset.
Collapse
Affiliation(s)
- Joshua D. Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Sarah E. Wolfe
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Amber R. Stewart
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Erika Tiemeier
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Sara E. Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Mayra Bueno Guerrero
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
3
|
The Effects of GABAergic System under Cerebral Ischemia: Spotlight on Cognitive Function. Neural Plast 2020; 2020:8856722. [PMID: 33061952 PMCID: PMC7539123 DOI: 10.1155/2020/8856722] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022] Open
Abstract
In this review, we present evidence about the changes of the GABAergic system on the hippocampus under the ischemic environment, which may be an underlying mechanism to the ischemia-induced cognitive deficit. GABAergic system, in contrast to the glutamatergic system, is considered to play an inhibitory effect on the central nervous system over the past several decades. It has received widespread attention in the area of schizophrenia and epilepsy. The GABAergic system has a significant effect in promoting neural development and formation of local neural circuits of the brain, which is the structural basis of cognitive function. There have been a number of reviews describing changes in the GABAergic system in cerebral ischemia in recent years. However, no study has investigated the changes in the system in the hippocampus during cerebral ischemic injury, which results in cognitive impairment, particularly at the chronic ischemic stage and the late phase of ischemia. We present a review of the changes of the GABAergic system in the hippocampus under ischemia, including GABA interneurons, extracellular GABA neurotransmitter, and GABA receptors. Several studies are also listed correlating amelioration of cognitive impairment by regulating the GABAergic system in the hippocampus damaged under ischemia. Furthermore, exogenous cell transplantation, which improves cognition by modulating the GABAergic system, will also be described in this review to bring new insight and strategy on solving cognitive deficits caused by cerebral ischemia.
Collapse
|
4
|
Kim DH, Kwon H, Choi JW, Shin CY, Cheong JH, Park SJ, Ryu JH. Roles of GABA A receptor α5 subunit on locomotion and working memory in transient forebrain ischemia in mice. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109962. [PMID: 32428535 DOI: 10.1016/j.pnpbp.2020.109962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
The γ-aminobutyric acid A (GABAA) receptor, which contains a chloride channel, is a typical inhibitory neurotransmitter receptor in the central nervous system. Although the GABAergic neurotransmitter system has been discovered to be involved in various psychological behaviors, such as anxiety, convulsions, and cognitive function, its functional changes under conditions of ischemic pathological situation are still uncovered. In the present study, we attempted to elucidate the functional changes in the GABAergic system after transient forebrain ischemia in mice. A bilateral common carotid artery occlusion for 20 min was used to establish a model of transient forebrain ischemia/reperfusion (tI/R). Delayed treatment with diazepam, a positive allosteric modulator of the GABAA receptor, increased locomotor activity in the open field test and spontaneous alternations in the Y-maze test in tI/R mice, but not in shams. Delayed treatment with diazepam did not alter neuronal death or the number of GABAergic neurons in tI/R mice. However, tI/R induced changes in the protein levels of GABAA receptor subunits in the hippocampus. In particular, the most marked increase in the tI/R group was found in the level of α5 subunit of the GABAA receptor. Similar to delayed treatment with diazepam, delayed treatment with imidazenil, an α5-sensitive benzodiazepine, increased spontaneous alternations in the Y-maze in tI/R mice, whereas zolpidem, an α5-insensitive benzodiazepine, failed to show such effects. These results suggest that tI/R-induced changes in the level of the α5 subunit of the GABAA receptor can alter the function of GABAergic drugs in a mouse model of forebrain ischemia.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Huiyoung Kwon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea.
| | - Ji Woong Choi
- Laboratory of Neuropharmacology, College of Pharmacy, Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea.
| | - Chan Young Shin
- Departments of Pharmacology and Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea.
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea.
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Sciences, Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
5
|
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: 29] [Impact Index Per Article: 5.8] [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
|
6
|
PKC and CaMK-II inhibitions coordinately rescue ischemia-induced GABAergic neuron dysfunction. Oncotarget 2018; 8:39309-39322. [PMID: 28445148 PMCID: PMC5503615 DOI: 10.18632/oncotarget.16947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/15/2017] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia leads to neuronal death for stroke, in which the imbalance between glutamatergic neurons and GABAergic neurons toward neural excitotoxicity is presumably involved. GABAergic neurons are vulnerable to pathological factors and impaired in an early stage of ischemia. The rescue of GABAergic neurons is expected to be the strategy to reserve ischemic neuronal impairment. As protein kinase C (PKC) and calmodulin-dependent protein kinase II (CaMK-II) are activated during ischemia, we have investigated whether the inhibitions of these kinases rescue the ischemic impairment of cortical GABAergic neurons. The functions of GABAergic neurons were analyzed by whole-cell recording in the cortical slices during ischemia and in presence of 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine (CaMK-II inhibitor) and chelerythrine chloride (PKC inhibitor). Our results indicate that PKC inhibitor or CaMK-II inhibitor partially prevents ischemia-induced functional deficits of cortical GABAergic neurons. Moreover, the combination of PKC and CaMK-II inhibitors synergistically reverses this ischemia-induced deficit of GABAergic neurons. One of potential therapeutic strategies for ischemic stroke may be to rescue the ischemia-induced deficit of cortical GABAergic neurons by inhibiting PKC and CaMK-II.
Collapse
|
7
|
Liang W, Huang X, Chen W. The Effects of Baicalin and Baicalein on Cerebral Ischemia: A Review. Aging Dis 2017; 8:850-867. [PMID: 29344420 PMCID: PMC5758355 DOI: 10.14336/ad.2017.0829] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke, producing a high mortality and morbidity rate, is a common clinical disease. Enhancing the prevention and control of ischemic stroke is particularly important. Baicalin and its aglycon baicalein are flavonoids extracted from Scutellaria baicalensis, an important traditional Chinese herb. In recent years, a growing body of evidences has shown that baicalin and baicalein could be effective in the treatment of cerebral ischemia. Pharmacokinetic studies have shown that baicalin could penetrate the blood-brain barrier and distribute in cerebral nuclei. Through a variety of in vitro and in vivo models of ischemic neuronal injury, numerous studies have demonstrated that baicalin and baicalein have salutary effect for neuroprotection. Especially, the studies on the pharmacological mechanism showed that baicalin and baicalein have several pharmacological activities, which include antioxidant, anti-apoptotic, anti-inflammatory and anti-excitotoxicity effects, protection of the mitochondria, promoting neuronal protective factors expression and adult neurogenesis effects and many more. This review focuses on the neuroprotective effects of baicalin and baicalein in ischemia or stroke-induced neuronal cell death. We aimed at collecting all important information regarding the neuroprotective effect and its pharmacological mechanism of baicalin and baicalein in various in vivo and in vitro experimental models of ischemic neuronal injury.
Collapse
Affiliation(s)
- Wei Liang
- Department of Traditional Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaobo Huang
- Department of Traditional Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wenqiang Chen
- Department of Traditional Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, ChinaThese authors equally contributed to this work
| |
Collapse
|
8
|
Liu Z, Huang Y, Liu L, Zhang L. Inhibitions of PKC and CaMK-II synergistically rescue ischemia-induced astrocytic dysfunction. Neurosci Lett 2017; 657:199-203. [DOI: 10.1016/j.neulet.2017.08.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 01/29/2023]
|
9
|
Carron SF, Alwis DS, Rajan R. Traumatic Brain Injury and Neuronal Functionality Changes in Sensory Cortex. Front Syst Neurosci 2016; 10:47. [PMID: 27313514 PMCID: PMC4889613 DOI: 10.3389/fnsys.2016.00047] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 05/19/2016] [Indexed: 01/21/2023] Open
Abstract
Traumatic brain injury (TBI), caused by direct blows to the head or inertial forces during relative head-brain movement, can result in long-lasting cognitive and motor deficits which can be particularly consequential when they occur in young people with a long life ahead. Much is known of the molecular and anatomical changes produced in TBI but much less is known of the consequences of these changes to neuronal functionality, especially in the cortex. Given that much of our interior and exterior lives are dependent on responsiveness to information from and about the world around us, we have hypothesized that a significant contributor to the cognitive and motor deficits seen after TBI could be changes in sensory processing. To explore this hypothesis, and to develop a model test system of the changes in neuronal functionality caused by TBI, we have examined neuronal encoding of simple and complex sensory input in the rat’s exploratory and discriminative tactile system, the large face macrovibrissae, which feeds to the so-called “barrel cortex” of somatosensory cortex. In this review we describe the short-term and long-term changes in the barrel cortex encoding of whisker motion modeling naturalistic whisker movement undertaken by rats engaged in a variety of tasks. We demonstrate that the most common form of TBI results in persistent neuronal hyperexcitation specifically in the upper cortical layers, likely due to changes in inhibition. We describe the types of cortical inhibitory neurons and their roles and how selective effects on some of these could produce the particular forms of neuronal encoding changes described in TBI, and then generalize to compare the effects on inhibition seen in other forms of brain injury. From these findings we make specific predictions as to how non-invasive extra-cranial electrophysiology can be used to provide the high-precision information needed to monitor and understand the temporal evolution of changes in neuronal functionality in humans suffering TBI. Such detailed understanding of the specific changes in an individual patient’s cortex can allow for treatment to be tailored to the neuronal changes in that particular patient’s brain in TBI, a precision that is currently unavailable with any technique.
Collapse
Affiliation(s)
- Simone F Carron
- Neuroscience Research Program, Biomedicine Discovery Institute, Department of Physiology, Monash University Monash, VIC, Australia
| | - Dasuni S Alwis
- Neuroscience Research Program, Biomedicine Discovery Institute, Department of Physiology, Monash University Monash, VIC, Australia
| | - Ramesh Rajan
- Neuroscience Research Program, Biomedicine Discovery Institute, Department of Physiology, Monash UniversityMonash, VIC, Australia; Ear Sciences Institute of AustraliaPerth, WA, Australia
| |
Collapse
|
10
|
Abstract
Decreased oxygen availability impairs cellular energy production and, without a coordinated and matched decrease in energy consumption, cellular and whole organism death rapidly ensues. Of particular interest are mechanisms that protect brain from low oxygen injury, as this organ is not only the most sensitive to hypoxia, but must also remain active and functional during low oxygen stress. As a result of natural selective pressures, some species have evolved molecular and physiological mechanisms to tolerate prolonged hypoxia with no apparent detriment. Among these mechanisms are a handful of responses that are essential for hypoxia tolerance, including (i) sensors that detect changes in oxygen availability and initiate protective responses; (ii) mechanisms of energy conservation; (iii) maintenance of basic brain function; and (iv) avoidance of catastrophic cell death cascades. As the study of hypoxia-tolerant brain progresses, it is becoming increasingly apparent that mitochondria play a central role in regulating all of these critical mechanisms. Furthermore, modulation of mitochondrial function to mimic endogenous neuroprotective mechanisms found in hypoxia-tolerant species confers protection against otherwise lethal hypoxic stresses in hypoxia-intolerant organs and organisms. Therefore, lessons gleaned from the investigation of endogenous mechanisms of hypoxia tolerance in hypoxia-tolerant organisms may provide insight into clinical pathologies related to low oxygen stress.
Collapse
Affiliation(s)
- Matthew E. Pamenter
- Department of Zoology, The University of British Columbia, #4200-6270 University Boulevard, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
11
|
Dai J, Chen L, Qiu YM, Li SQ, Xiong WH, Yin YH, Jia F, Jiang JY. Activations of GABAergic signaling, HSP70 and MAPK cascades are involved in baicalin's neuroprotection against gerbil global ischemia/reperfusion injury. Brain Res Bull 2013; 90:1-9. [PMID: 23041106 DOI: 10.1016/j.brainresbull.2012.09.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 09/19/2012] [Accepted: 09/25/2012] [Indexed: 02/06/2023]
Abstract
Baicalin, a flavonoid compound isolated from the plant Scutellaria baicalensis Georgi, is known as a protective agent against delayed neuronal cell death after ischemia/reperfusion. To investigate the neuroprotective mechanism of baicalin, the present study was conducted to explore whether the alterations of GABAergic signaling, heat shock protein 70 (HSP70) and mitogen-activated protein kinases (MAPKs) were involved in its neuroprotection on gerbils global ischemia. The bilateral carotid arteries were occluded by 5 min and baicalin at the dose of 200 mg/kg was intraperitoneally injected into the gerbils immediately after cerebral ischemia. Seven days after reperfusion, neurological deficit was scored and changes in hippocampal neuronal cell death were assessed by Nissl staining as well as NeuN immunohistochemistry. The mRNA and protein expressions of GABAergic signal molecules (GABA(A)R α1, GABA(A)R γ2, KCC2 and NKCC1) were determined in ischemic hippocampus by real-time RT-PCR and Western blot, respectively. In addition, HSP70 and MAPKs cascades (ERK, JNK and p38) were also detected using western blot assay. Our results illustrated that baicalin treatment significantly facilitated neurological function, suppressed the ischemia-induced neuronal damage. Besides, administration of baicalin also caused a striking increase of GABA(A)R α1, GABA(A)R γ2 and KCC2 together with the decrease of NKCC1 at mRNA and protein levels in gerbils hippocampus following an ischemic insult. Furthermore, the protein expressions of HSP70 and phosphorylated ERK (p-ERK) were evidently augmented while the phosphorylated JNK (p-JNK) and phosphorylated p38 (p-p38) were strikingly diminished in ischemic gerbils with baicalin treatment. These findings suggest that baicalin activates GABAergic signaling, HSP70 and MAPKs cascades in global ischemia, which may be a mechanism underlying the baicalin's neuroprotection.
Collapse
Affiliation(s)
- Jiong Dai
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Kelley MH, Ortiz J, Shimizu K, Grewal H, Quillinan N, Herson PS. Alterations in Purkinje cell GABAA receptor pharmacology following oxygen and glucose deprivation and cerebral ischemia reveal novel contribution of β1 -subunit-containing receptors. Eur J Neurosci 2012; 37:555-63. [PMID: 23176253 DOI: 10.1111/ejn.12064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 10/23/2012] [Accepted: 10/25/2012] [Indexed: 11/26/2022]
Abstract
Cerebellar Purkinje cells (PCs) are particularly sensitive to cerebral ischemia, and decreased GABA(A) receptor function following injury is thought to contribute to PC sensitivity to ischemia-induced excitotoxicity. Here we examined the functional properties of the GABA(A) receptors that are spared following ischemia in cultured Purkinje cells from rat and in vivo ischemia in mouse. Using subunit-specific positive modulators of GABA(A) receptors, we observed that oxygen and glucose deprivation (OGD) and cardiac arrest-induced cerebral ischemia cause a decrease in sensitivity to the β(2/3) -subunit-preferring compound, etomidate. However, sensitivity to propofol, a β-subunit-acting compound that modulates β(1-3) -subunits, was not affected by OGD. The α/γ-subunit-acting compounds, diazepam and zolpidem, were also unaffected by OGD. We performed single-cell reverse transcription-polymerase chain reaction on isolated PCs from acutely dissociated cerebellar tissue and observed that PCs expressed the β(1) -subunit, contrary to previous reports examining GABA(A) receptor subunit expression in PCs. GABA(A) receptor β(1) -subunit protein was also detected in cultured PCs by western blot and by immunohistochemistry in the adult mouse cerebellum and levels remained unaffected by ischemia. High concentrations of loreclezole (30 μm) inhibited PC GABA-mediated currents, as previously demonstrated with β(1) -subunit-containing GABA(A) receptors expressed in heterologous systems. From our data we conclude that PCs express the β(1) -subunit and that there is a greater contribution of β(1) -subunit-containing GABA(A) receptors following OGD.
Collapse
Affiliation(s)
- Melissa H Kelley
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | | | | | | | | | | |
Collapse
|
13
|
Montori S, DosAnjos S, Poole A, Regueiro-Purriños MM, Llorente IL, Darlison MG, Fernández-López A, Martínez-Villayandre B. Differential effect of transient global ischaemia on the levels of γ-aminobutyric acid type A (GABAA) receptor subunit mRNAs in young and older rats. Neuropathol Appl Neurobiol 2012; 38:710-22. [DOI: 10.1111/j.1365-2990.2012.01254.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
14
|
Desrues L, Lefebvre T, Lecointre C, Schouft MT, Leprince J, Compère V, Morin F, Proust F, Gandolfo P, Tonon MC, Castel H. Down-regulation of GABA(A) receptor via promiscuity with the vasoactive peptide urotensin II receptor. Potential involvement in astrocyte plasticity. PLoS One 2012; 7:e36319. [PMID: 22563490 PMCID: PMC3341351 DOI: 10.1371/journal.pone.0036319] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 04/02/2012] [Indexed: 02/07/2023] Open
Abstract
GABAA receptor (GABAAR) expression level is inversely correlated with the proliferation rate of astrocytes after stroke or during malignancy of astrocytoma, leading to the hypothesis that GABAAR expression/activation may work as a cell proliferation repressor. A number of vasoactive peptides exhibit the potential to modulate astrocyte proliferation, and the question whether these mechanisms may imply alteration in GABAAR-mediated functions and/or plasma membrane densities is open. The peptide urotensin II (UII) activates a G protein-coupled receptor named UT, and mediates potent vasoconstriction or vasodilation in mammalian vasculature. We have previously demonstrated that UII activates a PLC/PIPs/Ca2+ transduction pathway, via both Gq and Gi/o proteins and stimulates astrocyte proliferation in culture. It was also shown that UT/Gq/IP3 coupling is regulated by the GABAAR in rat cultured astrocytes. Here we report that UT and GABAAR are co-expressed in cerebellar glial cells from rat brain slices, in human native astrocytes and in glioma cell line, and that UII inhibited the GABAergic activity in rat cultured astrocytes. In CHO cell line co-expressing human UT and combinations of GABAAR subunits, UII markedly depressed the GABA current (β3γ2>α2β3γ2>α2β1γ2). This effect, characterized by a fast short-term inhibition followed by drastic and irreversible run-down, is not relayed by G proteins. The run-down partially involves Ca2+ and phosphorylation processes, requires dynamin, and results from GABAAR internalization. Thus, activation of the vasoactive G protein-coupled receptor UT triggers functional inhibition and endocytosis of GABAAR in CHO and human astrocytes, via its receptor C-terminus. This UII-induced disappearance of the repressor activity of GABAAR, may play a key role in the initiation of astrocyte proliferation.
Collapse
Affiliation(s)
- Laurence Desrues
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Thomas Lefebvre
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Céline Lecointre
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Marie-Thérèse Schouft
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Jérôme Leprince
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Vincent Compère
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France
| | - Fabrice Morin
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - François Proust
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- Department of Neurosurgery, Rouen University Hospital, Rouen, France
| | - Pierrick Gandolfo
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Marie-Christine Tonon
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Hélène Castel
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- * E-mail:
| |
Collapse
|
15
|
Oxygen Sensitive Synaptic Neurotransmission in Anoxia-Tolerant Turtle Cerebrocortex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 758:71-9. [DOI: 10.1007/978-94-007-4584-1_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
16
|
Mao X, Ji C, Sun C, Cao D, Ma P, Ji Z, Cao F, Min D, Li S, Cai J, Cao Y. Topiramate attenuates cerebral ischemia/reperfusion injury in gerbils via activating GABAergic signaling and inhibiting astrogliosis. Neurochem Int 2011; 60:39-46. [PMID: 22085431 DOI: 10.1016/j.neuint.2011.10.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 10/12/2011] [Accepted: 10/28/2011] [Indexed: 11/30/2022]
Abstract
Impaired GABAergic inhibitory synaptic transmission plays an essential role in the pathogenesis of selective neuronal cell death following transient global ischemia. GABA(A) receptor (GABA(A)R), K⁺-Cl⁻ co-transporter 2 (KCC2), Na⁺-K⁺-Cl⁻ co-transporter 1 (NKCC1) and astrocytes are of particular importance to GABAergic transmission. The present study was designed to explore whether the neuroprotective effect of topiramate (TPM) was linked with the alterations of GABAergic signaling and astrocytes. The bilateral carotid arteries were occluded, and TPM (80 mg/kg/day (divided twice daily), i.p.) was injected into gerbils. At day 1, 3 and 7 post-ischemia, neurological deficit was scored and changes in hippocampal neuronal cell death were evaluated by Nissl staining. The apoptosis-related regulatory proteins (procaspase-3, caspase-3, Bax and Bcl-2) and GABAergic signal molecules (GABA(A)R α1, GABA(A)R γ2, KCC2 and NKCC1) were also detected using western blot assay. In addition, the fluorescent intensity and protein level of glial fibrillary acidic protein (GFAP), a major component of astrocyte, were examined by confocal and immunoblot analysis. Our results showed that TPM treatment significantly decreased neurological deficit scores, attenuated the ischemia-induced neuronal loss and remarkably decreased the expression levels of procaspase-3, caspase-3 as well as the ratio of Bax/Bcl-2. Besides, treatment with TPM also resulted in the increased protein expressions of GABA(A)R α1, GABA(A)R γ2 and KCC2 together with the decreased protein level of NKCC1 in gerbils hippocampus. Furthermore, fluorescent intensity and protein level of GFAP were evidently reduced in TPM-treated gerbils. These findings suggest that the therapeutic effect of TPM on global ischemia/reperfusion injury appears to be associated with the enhancement of GABAergic signaling and the inhibition of astrogliosis in gerbils.
Collapse
Affiliation(s)
- Xiaoyuan Mao
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ujita S, Mizunuma M, Matsuki N, Ikegaya Y. Asynchronously enhanced spiking activity of ischemic neuronal networks. Biol Pharm Bull 2011; 34:764-7. [PMID: 21532170 DOI: 10.1248/bpb.34.764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cerebral ischemia causes the depletion of oxygen and nutrition from brain tissues, and when persistent, results in irreversible damage to the cell function and survival. The cellular response to ischemic conditions and its mechanisms have been investigated widely in in vivo and in vitro experimental models, yet no study has addressed the response of a whole neuronal network to energy deprivation with the single-cell resolution. Observations at the level of network are necessary, because the activity of individual neurons is nonlinearly integrated through a network and thereby gives rise to unexpectedly complex dynamics. Here we used functional multineuron calcium imaging (fMCI), an optical recording technique with high temporal and spatial resolution, to visualize the activity of neuron populations in hippocampus CA1 region under ischemia-like conditions ex vivo. We found that, although neurons responded to oxygen and glucose deprivation with an increase in the event frequency, they maintained an asynchronous network state. This is in contrast with other well known pathological states, in which the network hyperexcitability is usually accompanied by an increase in synchrony. We suggest that under ischemic conditions, at least to some time point, the neuronal network maintains the excitatory and inhibitory balance as a whole, whether actively or as a consequence of the cellular response to energy deprivation.
Collapse
Affiliation(s)
- Sakiko Ujita
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
18
|
Pamenter ME, Hogg DW, Ormond J, Shin DS, Woodin MA, Buck LT. Endogenous GABA(A) and GABA(B) receptor-mediated electrical suppression is critical to neuronal anoxia tolerance. Proc Natl Acad Sci U S A 2011; 108:11274-9. [PMID: 21690381 PMCID: PMC3131309 DOI: 10.1073/pnas.1102429108] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Anoxic insults cause hyperexcitability and cell death in mammalian neurons. Conversely, in anoxia-tolerant turtle brain, spontaneous electrical activity is suppressed by anoxia (i.e., spike arrest; SA) and cell death does not occur. The mechanism(s) of SA is unknown but likely involves GABAergic synaptic transmission, because GABA concentration increases dramatically in anoxic turtle brain. We investigated this possibility in turtle cortical neurons exposed to anoxia and/or GABA(A/B) receptor (GABAR) modulators. Anoxia increased endogenous slow phasic GABAergic activity, and both anoxia and GABA reversibly induced SA by increasing GABA(A)R-mediated postsynaptic activity and Cl(-) conductance, which eliminated the Cl(-) driving force by depolarizing membrane potential (∼8 mV) to GABA receptor reversal potential (∼-81 mV), and dampened excitatory potentials via shunting inhibition. In addition, both anoxia and GABA decreased excitatory postsynaptic activity, likely via GABA(B)R-mediated inhibition of presynaptic glutamate release. In combination, these mechanisms increased the stimulation required to elicit an action potential >20-fold, and excitatory activity decreased >70% despite membrane potential depolarization. In contrast, anoxic neurons cotreated with GABA(A+B)R antagonists underwent seizure-like events, deleterious Ca(2+) influx, and cell death, a phenotype consistent with excitotoxic cell death in anoxic mammalian brain. We conclude that increased endogenous GABA release during anoxia mediates SA by activating an inhibitory postsynaptic shunt and inhibiting presynaptic glutamate release. This represents a natural adaptive mechanism in which to explore strategies to protect mammalian brain from low-oxygen insults.
Collapse
Affiliation(s)
- Matthew E. Pamenter
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92037
| | - David W. Hogg
- Department of Cell and Systems Biology and Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | - Jake Ormond
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, Canada T1K 3M4; and
| | - Damian S. Shin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208
| | - Melanie A. Woodin
- Department of Cell and Systems Biology and Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| | - Leslie T. Buck
- Department of Cell and Systems Biology and Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada M5S 3G5
| |
Collapse
|
19
|
Transient enhancement of inhibitory synaptic transmission in hippocampal CA1 pyramidal neurons after cerebral ischemia. Neuroscience 2009; 160:412-8. [PMID: 19258028 DOI: 10.1016/j.neuroscience.2009.02.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 02/16/2009] [Accepted: 02/19/2009] [Indexed: 12/22/2022]
Abstract
Pyramidal neurons in hippocampal CA1 regions are highly sensitive to cerebral ischemia. Alterations of excitatory and inhibitory synaptic transmission may contribute to the ischemia-induced neuronal degeneration. However, little is known about the changes of GABAergic synaptic transmission in the hippocampus following reperfusion. We examined the GABA(A) receptor-mediated inhibitory postsynaptic currents (IPSCs) in CA1 pyramidal neurons 12 and 24 h after transient forebrain ischemia in rats. The amplitudes of evoked inhibitory postsynaptic currents (eIPSCs) were increased significantly 12 h after ischemia and returned to control levels 24 h following reperfusion. The potentiation of eIPSCs was accompanied by an increase of miniature inhibitory postsynaptic current (mIPSC) amplitude, and an enhanced response to exogenous application of GABA, indicating the involvement of postsynaptic mechanisms. Furthermore, there was no obvious change of the paired-pulse ratio (PPR) of eIPSCs and the frequency of mIPSCs, suggesting that the potentiation of eIPSCs might not be due to the increased presynaptic release. Blockade of adenosine A1 receptors led to a decrease of eIPSCs amplitude in post-ischemic neurons but not in control neurons, without affecting the frequency of mIPSCs and the PPR of eIPSCs. Thus, tonic activation of adenosine A1 receptors might, at least in part, contribute to the enhancement of inhibitory synaptic transmission in CA1 neurons after forebrain ischemia. The transient enhancement of inhibitory neurotransmission might temporarily protect CA1 pyramidal neurons, and delay the process of neuronal death after cerebral ischemia.
Collapse
|
20
|
Kelley MH, Taguchi N, Ardeshiri A, Kuroiwa M, Hurn PD, Traystman RJ, Herson PS. Ischemic insult to cerebellar Purkinje cells causes diminished GABAA receptor function and allopregnanolone neuroprotection is associated with GABAA receptor stabilization. J Neurochem 2008; 107:668-78. [PMID: 18699862 DOI: 10.1111/j.1471-4159.2008.05617.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cerebellar Purkinje cells (PC) are particularly vulnerable to ischemic injury and excitotoxicity, although the molecular basis of this sensitivity remains unclear. We tested the hypothesis that ischemia causes rapid down-regulation of GABA(A) receptors in cerebellar PC, thereby increasing susceptibility to excitotoxicity. Oxygen-glucose deprivation (OGD) caused a decline in functional GABA(A) receptors, within the first hour of re-oxygenation. Decreased amplitude of miniature inhibitory post-synaptic potentials confirmed that OGD caused a significant decrease in functional synaptic GABA(A) receptors and quantitative Western blot analysis demonstrated the loss of GABA(A) receptor current was associated with a decline in total receptor protein. Interestingly, the potent neuroprotectant allopregnanolone (ALLO) prevented the decline in GABA(A) receptor current and protein. Consistent with our in vitro data, global ischemia in mice caused a significant decline in total cerebellar GABA(A) receptor protein and PC specific immunoreactivity. Moreover, ALLO provided strong protection of PC and prevented ischemia-induced decline in GABA(A) receptor protein. Our findings indicate that ischemia causes a rapid and sustained loss of GABA(A) receptors in PC, whereas ALLO prevents the decline in GABA(A) receptors and protects against ischemia-induced damage. Thus, interventions which prevent ischemia-induced decline in GABA(A) receptors may represent a novel neuroprotective strategy.
Collapse
Affiliation(s)
- Melissa H Kelley
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health & Science University, Portland, Oregon 97201, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang L, Greenfield LJ. Post-hypoxic changes in rat cortical neuron GABA A receptor function require L-type voltage-gated calcium channel activation. Neuropharmacology 2008; 56:198-207. [PMID: 18674547 DOI: 10.1016/j.neuropharm.2008.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 06/26/2008] [Accepted: 07/05/2008] [Indexed: 11/17/2022]
Abstract
Hypoxia modifies GABA(A) receptor (GABA(A)R) function and can cause seizures, encephalopathy or myoclonus. To characterize the effects of hypoxia on neuronal GABA(A)Rs, we subjected rat cortical neurons to 1% O2 for 2, 4 or 8h, followed by recovery times of 0-96h, and used whole-cell and perforated patch-clamp recording to assess GABA(A)R currents and pharmacology. Hypoxic exposure for 4h caused downregulation of maximal GABA current immediately following hypoxia and after 48h recovery without changing the EC50 for GABA. Two- and eight-hour hypoxic exposures had inconsistent effects on GABA(A)R currents. Maximal diazepam potentiation was increased immediately following 4h hypoxia, while potentiation by zolpidem was increased after 48h recovery. Pentobarbital enhancement and zinc inhibition of GABA currents were unchanged. Hypoxia also caused a depolarizing shift in the reversal potential of GABA-induced Cl(-) currents after 24h recovery. The L-type voltage-gated calcium channel (L-VGCC) blocker, nitrendipine, during hypoxia or control treatment prevented the reduction in GABA(A)R currents, and increased control currents over baseline. Nitrendipine also prevented the increase in zolpidem potentiation 48h after hypoxia, and blocked the depolarizing shift in Cl(-) reversal potential 24h after hypoxia. The effects of hypoxia on maximal GABA(A)R currents, zolpidem pharmacology and Cl(-) reversal potential thus require depolarization-induced calcium entry via L-VGCCs, and constitutive L-VGCC activity appears to reduce maximal GABA(A)R currents in control neurons via a calcium-dependent process. Calcium-dependent modulation of GABA(A)R currents via L-VGCCs may be a fundamental regulatory mechanism for GABA receptor function.
Collapse
Affiliation(s)
- Liping Wang
- Department of Neurology, University of Toledo College of Medicine, Health Science Campus, Toledo, OH 43614-2598, USA
| | | |
Collapse
|
22
|
Tateishi N, Shimoda T, Manako JI, Katsumata S, Shinagawa R, Ohno H. Relevance of astrocytic activation to reductions of astrocytic GABAA receptors. Brain Res 2006; 1089:79-91. [PMID: 16643860 DOI: 10.1016/j.brainres.2006.02.139] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 02/15/2006] [Accepted: 02/24/2006] [Indexed: 11/22/2022]
Abstract
Although astrocytes express gamma-aminobutyric acid subtype-A (GABAA) receptors in the mature brain, GABAA receptor expression in a cultivation state remains controversial. In this study, we investigated the alteration of astrocytic GABAA receptor expression in in vitro and in vivo studies to elucidate the relevance of astrocytic activation to reductions of astrocytic GABAA receptors. The GABA-evoked Cl- current (GABAA response) in cultured astrocytes was determined by recording in the whole-cell mode using a conventional patch-clamp technique under voltage-clamp conditions. The respective amplitudes of GABAA responses on days in vitro 1, 3-5, 7-10, and 12-15 were 1019+/-97, 512+/-76, 84+/-21, and 22+/-9 pA, respectively, suggesting that the GABAA response subsequently diminished with in vitro aging. In immunohistochemical and biochemical analyses, the expression of GABAA receptor beta-subunit decreased, whereas expressions of glial fibrillary acidic protein (GFAP) and S100B, hallmarks of astrocytic activation, increased dramatically in the cultured astrocytes with in vitro aging. With the use of [3H]SR95531, a GABAA-specific ligand, at 24 h after transient focal ischemia, binding was significantly reduced in the astrocytic fractions without affecting the synaptosomal fractions, and decreases in the mRNA expression level of GABAA receptor beta-subunits were concurrently observed. Interestingly, the loss of GABAA response in cultured astrocytes was mitigated by co-culturing with neurons or treatments with monoclonal S100B antibodies. These results indicate that astrocytic GABAA receptors are reduced with in vitro aging and cerebral ischemia, presumably through the overproduction of S100B in activated astrocytes.
Collapse
Affiliation(s)
- Narito Tateishi
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan.
| | | | | | | | | | | |
Collapse
|
23
|
Zhan RZ, Nadler JV, Schwartz-Bloom RD. Depressed responses to applied and synaptically-released GABA in CA1 pyramidal cells, but not in CA1 interneurons, after transient forebrain ischemia. J Cereb Blood Flow Metab 2006; 26:112-24. [PMID: 15959457 DOI: 10.1038/sj.jcbfm.9600171] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transient cerebral ischemia kills CA1 pyramidal cells of the hippocampus, whereas most CA1 interneurons survive. It has been proposed that calcium-binding proteins, neurotrophins, and/or inhibitory neuropeptides protect interneurons from ischemia. However, different synaptic responses early after reperfusion could also underlie the relative vulnerabilities to ischemia of pyramidal cells and interneurons. In this study, we used gramicidin perforated patch recording in ex vivo slices to investigate gamma-aminobutyric acid (GABA) synaptic function in CA1 pyramidal cells and interneurons 4 h after a bilateral carotid occlusion accompanied by hypovolemic hypotension. At this survival time, the amplitudes of both miniature inhibitory postsynaptic currents (mIPSCs) and GABA-evoked currents were reduced in CA1 pyramidal cells, but not in CA1 interneurons. In addition, the mean rise time of mIPSCs was reduced in pyramidal cells. The reversal potential for the GABA current (E(GABA)) did not shift toward depolarizing values in either cell type, indicating that the driving force for chloride was unchanged at this survival time. We conclude that early during reperfusion GABAergic neurotransmission is attenuated exclusively in pyramidal neurons. This is likely explained by reduced GABAA receptor sensitivity or clustering and possibly also reduced GABA release, rather than by an elevation of intracellular chloride. Impaired GABA function may contribute to ischemic neuronal death by enhancing the excitability of CA1 pyramidal cells and facilitating N-methyl-D-aspartic acid channel opening. Therefore, normalizing GABAergic function might be a useful pharmacological approach to counter excessive, and potentially excitotoxic, glutamatergic activity during the postischemic period.
Collapse
Affiliation(s)
- Ren-Zhi Zhan
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
24
|
Zhao P, Qian H, Xia Y. GABA and glycine are protective to mature but toxic to immature rat cortical neurons under hypoxia. Eur J Neurosci 2005; 22:289-300. [PMID: 16045482 DOI: 10.1111/j.1460-9568.2005.04222.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although recent studies suggest that gamma-aminobutyric acid (GABA) and glycine may be 'inhibitory' to mature neurons, but 'excitatory' to immature neurons under normoxia, it is unknown whether inhibitory neurotransmitters are differentially involved in neuronal response to hypoxia in immature and mature neurons. In the present study, we exposed rat cortical neurons to hypoxia (1% O2) and examined the effects of three major inhibitory neurotransmitters (GABA, glycine and taurine) on the hypoxic neurons at different neuronal ages [days in vitro (DIV)4-20]. Our data showed that the cortical neurons expressed both GABA(A) and glycine receptors with differential developmental profiles. GABA (10-2000 microm) was neuroprotective to hypoxic neurons of DIV20, but enhanced hypoxic injury in neurons of <DIV20. Glycine at low concentrations (10-100 microm) exhibited a similar pattern to GABA. However, higher concentrations of glycine (1000-2000 microm) for long-term exposure (48-72 h) displayed neuroprotection at all ages (DIV4-20). Taurine (10-2000 microm), unlike GABA and glycine, displayed protection only in DIV4 neurons, and was slightly toxic to neurons>DIV4. In comparison with delta-opioid receptor (DOR)-induced protection in DIV20 neurons exposed to 72 h of hypoxia, glycine-induced protection was weaker than that of DOR but stronger than that of GABA and taurine. These data suggest that the effects of the inhibitory neurotransmitters on hypoxic cortical neurons are age-dependent, with GABA and glycine being neurotoxic to immature neurons and neuroprotective to mature neurons.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, LMP 3107, New Haven, CT 06520, USA
| | | | | |
Collapse
|
25
|
Gilby KL, Sydserff SG, Robertson HA. Differential neuroprotective effects for three GABA-potentiating compounds in a model of hypoxia–ischemia. Brain Res 2005; 1035:196-205. [PMID: 15722059 DOI: 10.1016/j.brainres.2004.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Revised: 11/30/2004] [Accepted: 12/04/2004] [Indexed: 10/25/2022]
Abstract
Clomethiazole (CMZ) is a GABA(A)-potentiating compound; however, it is unclear whether this mode of action is responsible for its neuroprotective effects in animal models of ischemia. This study compared the neuroprotective efficacies of muscimol and midazolam, two potent GABA(A)-potentiating compounds, to that of CMZ in a model of hypoxia-ischemia (H-I). To establish a neuroprotective profile for CMZ, CMZ (60, 95, or 125 mg kg-1, i.p.) was administered to post-natal day 25 male rats at numerous post-hypoxic time points and the rats were sacrificed 1 or 4 weeks later. Varying degrees of histological protection were evident when CMZ was administered 1, 2, or 3 h post-hypoxia with the 125 mg kg-1 dose producing complete histological protection if administered 3 h post-hypoxia. To determine whether midazolam or muscimol could match the protection provided by CMZ administered 3 h post-hypoxia, H-I rats received varying doses of these compounds 3 h post-hypoxia and were sacrificed 1 week later. Under identical conditions, no dose of muscimol or midazolam provided equivalent neuroprotection to that provided by CMZ. In fact, muscimol showed no neuroprotective ability whatsoever. Thus, CMZ, administered as late as 3 h post-hypoxia, was able to completely prevent H-I-induced cell death while a full dose range of other GABA-potentiating agents did not. Such direct comparison of these compounds in this model suggests the mechanism underlying the protective effects of CMZ may not rely solely on GABA(A)-potentiating properties. Elucidation of a novel mechanism of action for CMZ may expose new therapeutic targets in stroke treatment.
Collapse
Affiliation(s)
- K L Gilby
- Laboratory of Molecular Neurobiology, Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4H7.
| | | | | |
Collapse
|
26
|
Mielke JG, Wang YT. Insulin exerts neuroprotection by counteracting the decrease in cell-surface GABAA receptors following oxygen-glucose deprivation in cultured cortical neurons. J Neurochem 2005; 92:103-13. [PMID: 15606900 DOI: 10.1111/j.1471-4159.2004.02841.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A loss of balance between excitatory and inhibitory signaling leads to excitoxicity, and contributes to ischemic cell death. Reduced synaptic inhibition as a result of dysfunction of the ionotropic GABAA receptor has been suggested as one of the major causes for this imbalance, although the underlying mechanisms remain poorly understood. In the present study, we investigated whether oxygen-glucose deprivation (OGD), an ischemia-like challenge, alters cell-surface expression of GABAA receptors in cultured hippocampal neurons, and thereby leads to excitotoxic cell death. Using cell culture ELISA as a cell surface receptor assay, we found that OGD produced a marked decrease in cell surface GABAA receptors, without altering the total amount of receptors. Furthermore, the reduction could be prevented by inhibition of receptor endocytosis with hypertonic sucrose treatment. Notably, insulin significantly limited OGD-induced changes in cell-surface GABAA receptors. In parallel, insulin protected cultured neurons against both glutamate toxicity and OGD, as assayed by mitochondrial reduction of Alamar Blue. Importantly, insulin-mediated neuroprotection was eliminated when bicuculline, a GABAA receptor antagonist, was co-applied with insulin during OGD. Together, our results strongly suggest that ischemia-like insults decrease cell surface GABAA receptors in neurons via accelerated internalization, and that insulin provides neuroprotection by counteracting this reduction.
Collapse
Affiliation(s)
- John G Mielke
- Brain and Behavior Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | |
Collapse
|
27
|
Gilby KL, Kelly ME, McIntyre DC, Robertson HA. Neuro-overprotection? A functional evaluation of clomethiazole-induced neuroprotection following hypoxic-ischemic injury. Neuroscience 2005; 131:785-92. [PMID: 15749333 DOI: 10.1016/j.neuroscience.2004.11.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2004] [Indexed: 10/25/2022]
Abstract
Hypoxic-ischemic (H-I) injury produces extensive damage to the hippocampus of young rats. We have recently shown that administration of 125 mg kg-1 clomethiazole (CMZ), a GABA(A)-agonist, provides complete histological protection against H-I injury if administered 3 h post-H-I (Brain Res 1035 (2005) 194). However, whether that histological protection translates into lasting functional preservation is unclear. To determine whether hippocampal-based circuits remain functionally intact in CMZ-protected H-I rats, we administered 125 mg kg-1 (high dose [CMZ-HD]) or 65 mg kg-1 (low dose [CMZ-LD]) CMZ, 3 h post-H-I, and examined numerous kindling parameters in the dorsal hippocampus 60 days following H-I. Kindling parameters included afterdischarge (AD) thresholds (ADTs), AD durations and kindling rates. Additional groups assessed included vehicle-injected H-I (VIH), hypoxic, ligated and naive rats. VIH, CMZ-HD, CMZ-LD and hypoxic rats all exhibited significantly faster kindling rates than naive rats. Thus, a previous traumatic event, even hypoxia alone, facilitated subsequent seizure propagation. Still, a significantly slower kindling rate was evident in CMZ-HD rats than in hypoxic, VIH or CMZ-LD rats. Moreover, while longer pre-kindling AD durations were observed in the damaged hippocampus of VIH compared with naive rats, this was not true for either CMZ-treated groups, hypoxic or ligated rats. Collectively, these findings suggest CMZ can suppress the epileptogenic effects of H-I. Surprisingly, however, both groups of CMZ-treated rats exhibited a four to nine times greater ADT than any other group and this effect was most profound in the CMZ-protected hippocampus. Thus, CMZ administration protected local neurons against terminal insult and left network excitability relatively normal with respect to seizure offset mechanisms but also caused profound elevation of local ADTs, which suggests a local hypoexcitability/increased inhibition. Finally, this study demonstrates, for the first time, that the kindling model can serve as a sensitive measure of function-related neuroprotective efficacy in animal models of ischemia.
Collapse
Affiliation(s)
- K L Gilby
- Laboratory of Molecular Neurobiology, Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4H7.
| | | | | | | |
Collapse
|
28
|
Steiger JL, Russek SJ. GABAA receptors: building the bridge between subunit mRNAs, their promoters, and cognate transcription factors. Pharmacol Ther 2004; 101:259-81. [PMID: 15031002 DOI: 10.1016/j.pharmthera.2003.12.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The type A gamma-aminobutyric acid (GABA(A)) receptors mediate the majority of fast inhibitory neurotransmission in the CNS, and alterations in GABA(A) receptor function is believed to be involved in the pathology of several neurological and psychiatric illnesses, such as epilepsy, anxiety, Alzheimer's disease, and schizophrenia. GABA(A) receptors can be assembled from eight distinct subunit families defined by sequence similarity: alpha(1-6), beta(1-3), gamma(1-3), delta, pi, theta, and rho(1-3). The regulation of GABA(A) receptor function in the brain is a highly compensating system, influencing both the number and the composition of receptors at the cell surface. While transcriptional and translational points of control operate in parallel, it is becoming increasingly evident that many functional changes in GABA(A) receptors reflect the differential gene regulation of its subunits. The fact that certain GABA(A) receptor subunit genes are transcribed in distinct cell types during specific periods of development strongly suggests that genetic control plays a major role in the choice of subunit variants available for receptor assembly. This review focuses on the physiological conditions that alter subunit mRNA levels, the promoters that may control such levels, and the use of a conceptual framework created by bioinformatics to study coordinate and independent GABA(A) receptor subunit gene regulation. As this exciting field moves closer to identifying the language hidden inside the chromatin of GABA(A) receptor subunit gene clusters, future experiments will be aimed at testing models generated by computational analysis with biologically relevant in vivo and in vitro assays. It is hoped that through this functional genomic approach there will be the identification of new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Janine L Steiger
- Laboratory of Molecular Neurobiology, Department of Pharmacology, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
29
|
Gao L, Lyons AR, Greenfield LJ. Hypoxia alters GABAA receptor function and subunit expression in NT2-N neurons. Neuropharmacology 2004; 46:318-30. [PMID: 14975687 DOI: 10.1016/j.neuropharm.2003.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2003] [Revised: 09/04/2003] [Accepted: 09/08/2003] [Indexed: 11/30/2022]
Abstract
Hypoxia causes dysfunction of excitatory and inhibitory neurotransmission, often resulting in encephalopathy, seizures or myoclonus. We evaluated the effects of hypoxia on GABAA receptor (GABAAR) function and expression in an in vitro model of neuronal hypoxia. NT2-N cells, derived from the human NT2 teratocarcinoma cell line, were exposed to < or =1% O2 for 8 h and then used immediately for experiments or allowed to recover under normoxic conditions (95% air/5% CO2) for 24, 48 or 96 h. Hypoxic treatment did not cause obvious morphological changes or cell death. In whole-cell patch-clamp recordings, the GABA current EC50 was unchanged, however, maximal GABA-evoked currents changed in a biphasic manner. Maximal GABA currents were significantly increased immediately after hypoxia, but were significantly reduced after 48 h normoxic recovery, and then returned to baseline after 96 h recovery. Maximal potentiation of 10 microM GABA currents by diazepam was increased 48 h after hypoxia, but potentiation by zolpidem was decreased. Barbiturate enhancement and zinc inhibition of GABA currents were unchanged. Semiquantitative reverse transcriptase (RT)-PCR showed decreased alpha1, alpha5, beta2 and gamma2 subunit mRNA after hypoxia. Hypoxic exposure altered GABAAR physiology and subunit mRNA expression, which may correlate with symptoms observed after hypoxia in vivo.
Collapse
Affiliation(s)
- Lei Gao
- Cellular and Molecular Neurobiology Program, Medical College of Ohio, 3120 Glendale Avenue, Ruppert Health Center, Suite 1450, Toledo, OH 43614, USA
| | | | | |
Collapse
|
30
|
Kim Y, Oh S. Changes of GABA(A) receptor binding and subunit mRNA level in rat brain by infusion of NOS inhibitor. Brain Res 2002; 952:246-56. [PMID: 12376186 DOI: 10.1016/s0006-8993(02)03248-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present study, we have investigated the effects of prolonged inhibition of nitric oxide synthase (NOS) by infusion of NOS inhibitor, L-nitroarginine, to examine the pentobarbital-induced sleep, modulation of GABA(A) receptor binding, and GABA(A) receptor subunit mRNA level in rat brain. Pre-treatment with L-nitroarginine 30 min before pentobarbital treatment (60 mg/kg, i.p.) significantly increased the duration of sleep in rats. However, the duration of pentobarbital-induced sleep was shortened by the prolonged infusion of L-nitroarginine into ventricle. We have investigated the effect of NOS inhibitor on GABA(A) receptor binding characteristics in discrete areas of brain regions by using autoradiographic and in situ hybridization techniques. Rats were infused with L-nitroarginine (10, 100 pmol/10 microl/h, i.c.v.) for 7 days, through pre-implanted cannula by osmotic minipumps. The levels of [(3)H]muscimol and [(3)H]flunitrazepam binding were markedly elevated in almost all of brain regions including cortex, caudate putamen, thalamus, hippocampus, and cerebellum. However, there was no change in the level of [(35)S]TBPS binding. The levels of beta2-subunit were elevated in the cortex, brainstem, and cerebellar granule layers. By contrast, the levels of beta3-subunit were significantly decreased in the cortex, hippocampus, and cerebellar granule layers in L-nitroarginine-infused rats. Following L-nitroarginine treatment, the levels of alpha6- and delta-subunits which were strictly localized to the cerebellum, were not changed in the cerebellar granule layer. These results show that the prolonged inhibition of NOS by L-nitroarginine-infusion markedly elevates [(3)H]muscimol and [(3)H]flunitrazepam binding throughout the brain, and alters GABA(A) receptor subunit mRNA levels in different directions. Chronic inhibition of NO generation has differential effects on the various expressions of GABA(A) receptor subunits. These suggest the involvement of different regulatory mechanisms for the NO-induced expression of GABA(A) receptor.
Collapse
Affiliation(s)
- Younghwa Kim
- Department of Anatomy, College of Medicine, Ewha Womans University, Seoul, South Korea
| | | |
Collapse
|
31
|
Sommer C, Fahrner A, Kiessling M. [3H]muscimol binding to gamma-aminobutyric acid(A) receptors is upregulated in CA1 neurons of the gerbil hippocampus in the ischemia-tolerant state. Stroke 2002; 33:1698-705. [PMID: 12053014 DOI: 10.1161/01.str.0000016404.14407.77] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Excitotoxic activation of glutamate receptors is currently thought to play a pivotal role in delayed neuronal death (DND) of highly vulnerable CA1 neurons in the gerbil hippocampus after transient global ischemia. Postischemic degeneration of these neurons can be prevented by "preconditioning" with a short sublethal ischemic stimulus. The present study was designed to test whether ischemic preconditioning is associated with specific alterations of ligand binding to excitatory glutamate and/or inhibitory gamma-aminobutyric acid (GABA)A receptors compared with ischemia severe enough to induce DND. METHODS With the use of quantitative receptor autoradiography, postischemic ligand binding of [3H]MK-801 and [3H]alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) to excitatory N-methyl-D-aspartate (NMDA) and AMPA receptors as well as [3H]muscimol to inhibitory GABA(A) receptors in hippocampal subfields CA1, CA3, and the dentate gyrus were analyzed in 2 experimental paradigms. Gerbils were subjected to (1) a 5-minute ischemic period resulting in DND of CA1 neurons and (2) a 2.5-minute period of ischemia mediating tolerance induction. RESULTS [3H]MK-801 and [3H]AMPA binding values to excitatory NMDA and AMPA receptors showed a delayed decrease in relatively ischemia-resistant CA3 and dentate gyrus despite maintained neuronal cell density. [3H]Muscimol binding to GABA(A) receptors in CA1 neurons was transiently but significantly increased after preconditioning but not after global ischemia with consecutive neuronal death. CONCLUSIONS Downregulation of ligand binding to glutamate receptors in relatively ischemia-resistant CA3 and dentate gyrus neurons destined to survive suggests marked synaptic reorganization processes despite maintained structural integrity. More importantly, upregulation of binding to inhibitory GABA(A) receptors in the hippocampus indicates a relative shift between inhibitory and excitatory neurotransmission that we suggest may participate in endogenous postischemic neuroprotection.
Collapse
Affiliation(s)
- Clemens Sommer
- Department of Neuropathology, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany.
| | | | | |
Collapse
|
32
|
Abstract
In this review, we present evidence for the role of gamma-aminobutyric acid (GABA) neurotransmission in cerebral ischemia-induced neuronal death. While glutamate neurotransmission has received widespread attention in this area of study, relatively few investigators have focused on the ischemia-induced alterations in inhibitory neurotransmission. We present a review of the effects of cerebral ischemia on pre and postsynaptic targets within the GABAergic synapse. Both in vitro and in vivo models of ischemia have been used to measure changes in GABA synthesis, release, reuptake, GABA(A) receptor expression and activity. Cellular events generated by ischemia that have been shown to alter GABA neurotransmission include changes in the Cl(-) gradient, reduction in ATP, increase in intracellular Ca(2+), generation of reactive oxygen species, and accumulation of arachidonic acid and eicosanoids. Neuroprotective strategies to increase GABA neurotransmission target both sides of the synapse as well, by preventing GABA reuptake and metabolism and increasing GABA(A) receptor activity with agonists and allosteric modulators. Some of these strategies are quite efficacious in animal models of cerebral ischemia, with sedation as the only unwanted side-effect. Based on promising animal data, clinical trials with GABAergic drugs are in progress for specific types of stroke. This review attempts to provide an understanding of the mechanisms by which GABA neurotransmission is sensitive to cerebral ischemia. Furthermore, we discuss how dysfunction of GABA neurotransmission may contribute to neuronal death and how neuronal death can be prevented by GABAergic drugs.
Collapse
Affiliation(s)
- R D Schwartz-Bloom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
| | | |
Collapse
|
33
|
Miyamoto O, Nakamura T, Yamagami S, Negi T, Tokuda M, Matsui H, Itano T. Depression of long term potentiation in gerbil hippocampus following postischemic hypothermia. Brain Res 2000; 873:168-72. [PMID: 10915827 DOI: 10.1016/s0006-8993(00)02521-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To investigate the mechanism of chronic cell death following postischemic hypothermia, the change of N-methyl-D-aspartate receptor (NMDAR) were examined by immunohistochemistry of NMDAR1 and long-term potentiation (LTP) in the CA1 subfield of the gerbil hippocampus. At 1 week following postischemic hypothermia (32 degrees Cx4 h), all CA1 neurons survived; however, immunoreactivity of NMDAR1 increased in neuronal perikarya whereas decreased in dendrites in the CA1 neurons. The abnormality was still observed in remaining CA1 neurons at 1 month after hypothermia. LTP was also significantly depressed at 1 week after hypothermia. These results suggest that some abnormalities in the glutamate receptor may be caused by ischemia; such abnormality would persist in spite of hypothermia treatment, resulting in the depression of LTP.
Collapse
Affiliation(s)
- O Miyamoto
- Department of Biology, Kagawa Medical University, Miki-cho, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Fedele E, Ansaldo MA, Varnier G, Raiteri M. Benzodiazepine-sensitive GABA(A) receptors limit the activity of the NMDA/NO/cyclic GMP pathway: a microdialysis study in the cerebellum of freely moving rats. J Neurochem 2000; 75:782-7. [PMID: 10899955 DOI: 10.1046/j.1471-4159.2000.0750782.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the cerebellum, infusion of NMDA (200 microM) for 20 min evoked a marked (200%) increase of extracellular cyclic GMP (cGMP) levels. The selective GABA(A) receptor agonist muscimol (0.01-100 microM) was able to counteract the NMDA effect with an EC(50) of 0.65 microM; the inhibitory effect of muscimol (10 microM) was prevented by bicuculline (50 microM). Diazepam (10 microM) significantly potentiated the muscimol (1 microM) inhibition; furthermore, when coinfused with 0.1 microM muscimol (a concentration not affecting, on its own, the cGMP response to NMDA), diazepam (10 microM) reduced the NMDA effect. Similar results were obtained with zolpidem (0.1-1 microM). Finally, local infusion of the benzodiazepine site antagonist flumazenil (10 microM), together with muscimol and diazepam, almost completely restored the effect of NMDA on extracellular cGMP levels. It is concluded that GABA(A) receptors potently control the NMDA/nitric oxide/cGMP pathway in the cerebellum in vivo. In terms of the alpha subunit composition, we can deduce that the cerebellar GABA(A) receptor does not contain alpha(6) or beta(4) subunits because it is diazepam-sensitive. Moreover, the observation that zolpidem is active at a rather low concentration, in combination with localization studies present in the literature, tend to exclude the presence of alpha(5) subunits in the receptor composition and suggest the involvement of an alpha(1) subunit.
Collapse
Affiliation(s)
- E Fedele
- Sezione di Farmacologia e Tossicologia, Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | | | | | | |
Collapse
|
35
|
Raghavendra Rao VL, Rao AM, Dogan A, Bowen KK, Hatcher J, Rothstein JD, Dempsey RJ. Glial glutamate transporter GLT-1 down-regulation precedes delayed neuronal death in gerbil hippocampus following transient global cerebral ischemia. Neurochem Int 2000; 36:531-7. [PMID: 10762090 DOI: 10.1016/s0197-0186(99)00153-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Glial (GLT-1 and GLAST) and neuronal (EAAC1) high-affinity transporters mediate the sodium dependent glutamate reuptake in mammalian brain. Their dysfunction leads to neuronal damage by allowing glutamate to remain in the synaptic cleft for a longer duration. The purpose of the present study is to understand their contribution to the ischemic delayed neuronal death seen in gerbil hippocampus following transient global cerebral ischemia. The protein levels of these three transporters were studied by immunoblotting as a function of reperfusion time (6 h to 7 days) following a 10 min occlusion of bilateral common carotid arteries in gerbils. In the vulnerable hippocampus, there was a significant decrease in the protein levels of GLT-1 (by 36-46%, P < 0.05; between 1 and 3 days of reperfusion) and EAAC1 (by 42-68%, P < 0.05; between 1 and 7 days of reperfusion). Histopathological evaluation showed no neuronal loss up to 2 days of reperfusion but an extensive neuronal loss (by approximately 84%, P < 0.01) at 7 days of reperfusion in the hippocampal CA1 region. The time frame of GLT-1 dysfunction (1-3 days of reperfusion) precedes the initiation of delayed neuronal death (2-3 days of reperfusion). This suggests GLT-1 dysfunction as a contributing factor for the hippocampal neuronal death following transient global cerebral ischemia. Furthermore, decreased EAAC1 levels may contribute to GABAergic dysfunction and excitatory/inhibitory imbalance following transient global ischemia.
Collapse
Affiliation(s)
- V L Raghavendra Rao
- Department of Neurological Surgery, University of Wisconsin-Madison, F4/309 Clinical Science Center, 53792, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
With the approval of alteplase (tPA) therapy for stroke, it is likely that combination therapy with tPA to restore blood flow, and agents like glutamate receptor antagonists to halt or reverse the cascade of neuronal damage, will dominate the future of stroke care. The authors describe events and potential targets of therapeutic intervention that contribute to the excitotoxic cascade underlying cerebral ischemic cell death. The focal and global animal models of stroke are the basis for the identification of these events and therapeutic targets. The signalling pathways contributing to ischemic neuronal death are discussed based on their cellular localization. Cell surface signalling events include the activities of both voltage-gated K+, Na+, and Ca2+ channels and ligand-gated glutamate, gamma-aminobutyric acid and adenosine receptors and channels. Intracellular signalling events include alterations in cytosolic and subcellular Ca2+ dynamics, Ca2+ -dependent kinases and immediate early genes whereas intercellular mechanisms include free radical formation and the activation of the immune system. An understanding of the relative importance and temporal sequence of these processes may result in an effective stroke therapy targeting several points in the cascade. The overall goal is to reduce disability and enhance quality of life for stroke survivors.
Collapse
Affiliation(s)
- D L Small
- Receptor and Ion Channels Group, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario.
| | | | | |
Collapse
|
37
|
Churn SB, DeLorenzo RJ. Modulation of GABAergic receptor binding by activation of calcium and calmodulin-dependent kinase II membrane phosphorylation. Brain Res 1998; 809:68-76. [PMID: 9795142 DOI: 10.1016/s0006-8993(98)00834-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
gamma-Aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system (CNS). Because of the important role that GABA plays in the CNS, alteration of GABAA receptor function would significantly affect neuronal excitability. Protein phosphorylation is a major mechanism for regulating receptor function in the brain and has been implicated in modulating GABAA receptor function. Therefore, this study was initiated to determine the role of calmodulin-dependent kinase II (CaM kinase II) membrane phosphorylation on GABAA receptor binding. Synaptosomal membrane fractions were tested for CaM kinase II activity towards endogenous substrates. In addition, muscimol binding was evaluated under equilibrium conditions in synaptosomal membrane fractions subjected to either basal (Mg2+ alone) or maximal CaM kinase II-dependent phosphorylation. Activation of endogenous CaM kinase II-dependent phosphorylation resulted in a significant enhancement of the apparent Bmax for muscimol binding without significantly altering the apparent binding affinity. The enhanced muscimol binding could be increased further by the addition of exogenous CaM kinase II to synaptosomal membrane fractions. Co-incubation with inhibitors of kinase activity during the phosphorylation reactions blocked the CaM kinase II-dependent increase in muscimol binding. The data support the hypothesis that activation of CaM kinase II-dependent phosphorylation caused an increased GABAA receptor binding and may play an important role in modulating the function of this inhibitory receptor/chloride ion channel complex.
Collapse
Affiliation(s)
- S B Churn
- Department of Neurology, Medical College of Virginia, Virginia Commonwealth University, Box 980599 MCV Station, Richmond, VA 23298-0599, USA.
| | | |
Collapse
|
38
|
Luhmann HJ, Raabe K, Qü M, Zilles K. Characterization of neuronal migration disorders in neocortical structures: extracellular in vitro recordings. Eur J Neurosci 1998; 10:3085-94. [PMID: 9786203 DOI: 10.1046/j.1460-9568.1998.00311.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The majority of patients showing neuronal migration disorders in cortical structures suffer from pharmaco-resistant epilepsy. In order to study the molecular and cellular mechanisms underlying this pronounced hyperexcitability, we used an animal model of focal cortical dysplasia demonstrating structural malformations which resemble the human pathology of microgyria. Neocortical slices prepared from adult rats, which at the day of birth received a cortical freeze lesion, were analysed in vitro with an array of eight extracellular recording electrodes to investigate the pattern and pharmacology of propagating epileptiform activity in microgyric cortex. In cortical slices exhibiting neuronal migration disorders, orthodromic synaptic stimulation elicited late recurrent activity and early epileptiform responses that spread with 0.06 m/s over > or = 3.5 mm across the cortex. Application of a N-methyl-D-aspartate (NMDA) antagonist blocked the late recurrent activity, but not the propagation of the early epileptiform responses. The latter were blocked by an (+/-)-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) antagonist, indicating that the spread of this activity was predominantly mediated by activation of AMPA receptors. A very similar response pattern could be observed in neocortical slices obtained from untreated age-matched control rats, when the slice was partially disinhibited by bath-application of 5 microM bicuculline methiodide. Stimulus-evoked epileptiform signals recorded in disinhibited slices propagated with 0.08 m/s across the cortex and showed the same sensitivity to ionotropic glutamate antagonists as in dysplastic cortex. Our results indicate that widespread structural and/or functional modifications of the AMPA receptor and possibly also of the gamma-amino-butyric acid type A receptor contribute to the pronounced hyperexcitability in dysplastic cortex.
Collapse
Affiliation(s)
- H J Luhmann
- Institute of Neurophysiology, University of Düsseldorf, Germany.
| | | | | | | |
Collapse
|
39
|
Zhang L, Rubinow DR, Ma W, Marks JM, Feldman AN, Barker JL, Tathan TA. GABA receptor subunit mRNA expression in brain of conflict, yoked control and control rats. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 58:16-26. [PMID: 9685573 DOI: 10.1016/s0169-328x(98)00061-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Animal conflict models have been used for years as a preclinical screen for predicting anxiolytic therapeutic efficacy. Anxiolytics, including benzodiazepines, increase punished responding. This suggests that the punished behavior may be mediated by the GABA receptor. To evaluate this hypothesis, we performed in situ hybridization histochemistry studies of GABA receptor subunits (alpha1-alpha4) and synthetic enzymes glutamic acid decarboxylase (GAD65 and GAD67) in four groups of rats: conflict (punishment), yoked controls (rats shocked without conflict training history), fixed interval only controls (rats that worked for food but were not shocked) and untreated controls. With conflict behavioral training, bilateral reduction of mRNA for the GABAA alpha1 subunit was seen relative to controls in the cortex, thalamus and hippocampus. In contrast, alteration of alpha2 mRNA levels appeared only in the yoked control group, with increased levels seen in the thalamus and cortex and decreased levels in the hippocampus. There were no differences in the alpha2 mRNA level between the control and the conflict behavioral trained animals. Further, no significant differences were found between groups in the mRNA levels for the alpha3 subunit, alpha4 subunit, GAD65, and GAD67. These results suggest that the behaviors related to conflict and uncontrollable aversive stimuli (yoked control group) are accompanied and perhaps mediated by selective changes in the GABAA alpha1 or alpha2 subunits, respectively. These findings highlight the potential usefulness of the conflict model as a means of elucidating the biological underpinnings of anxiety disorder. Published by Elsevier Science B.V. All rights reserved.
Collapse
Affiliation(s)
- L Zhang
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-1276, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Qü M, Mittmann T, Luhmann HJ, Schleicher A, Zilles K. Long-term changes of ionotropic glutamate and GABA receptors after unilateral permanent focal cerebral ischemia in the mouse brain. Neuroscience 1998; 85:29-43. [PMID: 9607700 DOI: 10.1016/s0306-4522(97)00656-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Long-term hyperexcitability was found after unilateral, permanent middle cerebral artery occlusion in exofocal neocortical areas of the adult mouse [Mittmann et al. (1998) Neuroscience 85, 15-27]. The aim of the present study was to test the hypothesis in an identical paradigm of ischemia. whether alterations in the densities of both excitatory and inhibitory amino acid receptors may underlie these pathophysiological changes. Alterations in densities of [3H]dizocilpine, [3H]D,L-amino-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, [3H]kainate and [3H]muscimol binding sites were demonstrated with quantitative in vitro receptor autoradiography. All binding sites were severely reduced in the core of the ischemic lesion. A completely different reaction was found in the exofocal, histologically inconspicuous parts of the somatosensory cortex and the more remote neocortical areas of both hemispheres. The [3H]muscimol binding sites were significantly reduced four weeks after ischemia in the motor cortex, hindlimb representation area and exofocal parts of the primary and secondary somatosensory cortices of both hemispheres. The focus of the reduction in [3H]muscimol binding sites was found in lower layer V and upper layer VI. Contrastingly, the densities of [3H]dizocilpine binding sites were found to be increased in these areas, whereas those of [3H]D,L-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and [3H]kainate binding sites did not show significant changes. The [3H]dizocilpine binding site density increased predominantly in layers III and IV. All binding sites were also reduced in the retrogradely reacting, gliotic part of the ipsilateral ventroposterior thalamic nucleus, whereas the [3H]D,L-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid binding sites were increased in the surround of the ipsilateral nucleus and no changes in binding sites were seen in the whole contralateral nucleus. We conclude that permanent local ischemia leads to a long-term and widespread impairment of the normal balance between binding sites of excitatory and inhibitory neurotransmitter receptors in neocortical areas far away from the focus of the post-ischemic tissue damage. The imbalance comprises an up-regulation of the [3H]dizocilpine binding sites in the ion channels of N-methyl-D-aspartate receptors and a down-regulation of [3H]muscimol binding sites of the GABA(A) receptors in the ipsi- and contralateral neocortex. These changes at the receptor level explain the previously observed hyperexcitability with the appearance of epileptiform field potentials and the long duration of excitatory postsynaptic potentials four weeks after ischemia.
Collapse
Affiliation(s)
- M Qü
- C. & O. Vogt Institute of Brain Research, University of Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
41
|
Mittmann T, Qü M, Zilles K, Luhmann HJ. Long-term cellular dysfunction after focal cerebral ischemia: in vitro analyses. Neuroscience 1998; 85:15-27. [PMID: 9607699 DOI: 10.1016/s0306-4522(97)00638-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The long-term (< or = six months) functional consequences of permanent middle cerebral artery occlusion were studied with in vitro extra- and intracellular recording techniques in adult mouse neocortical slices. After survival times of one to three days, 28 days and six months, intracellular recordings from layers II/III pyramidal cells in the vicinity of the infarct did not reveal any statistically significant changes in the intrinsic membrane properties when compared to age-matched control animals. However, a pronounced hyperexcitability could be observed upon orthodromic synaptic stimulation in neocortical slices obtained from mice 28 days after induction of ischemia. Low-intensity electrical stimulation of the afferents elicited particularly in this group epileptiform extracellular field potential responses and intracellular excitatory postsynaptic potentials, that were longer in duration as compared to the controls. When the N-methyl-D-aspartate receptor-mediated excitatory postsynaptic potential was pharmacologically isolated in a bathing solution containing 0.1 mM Mg2+ and 10 microM 6-cyano-7-nitroquinoxaline-2,3-dione, the synaptic responses were longer and larger in the ischemic cortex as compared to the controls. Higher stimulus intensities evoked in normal medium a biphasic inhibitory postsynaptic potential, that contained in the 28 days post-ischemia group a prominent amino-phosphonovaleric acid-sensitive component, indicating a strong concurrent activation of a N-methyl-D-aspartate receptor-mediated excitatory postsynaptic potential. This pronounced co-activation could only be observed in the 28 days ischemic group, and neither after one to three days or six months post-ischemia nor in the controls. The quantitative analysis of the efficiency of stimulus- evoked inhibitory postsynaptic potentials recorded in amino-phosphono-valeric acid revealed a reduction of GABA-mediated inhibition in ischemic cortex. Although this reduction in intracortical inhibition may already contribute to an augmentation of N-methyl-D-aspartate receptor-mediated excitation, our results do also indicate that the function of N-methyl-D-aspartate receptors is transiently enhanced in the ischemic cortex. This transient hyperexcitability does not only cause cellular dysfunction in the vicinity of the infarct, but may also contribute to neuronal damage due to excitotoxicity.
Collapse
Affiliation(s)
- T Mittmann
- Institute of Neurophysiology, University of Düsseldorf, Germany
| | | | | | | |
Collapse
|
42
|
Inglefield JR, Wilson CA, Schwartz-Bloom RD. Effect of transient cerebral ischemia on gamma-aminobutyric acidA receptor alpha 1-subunit-immunoreactive interneurons in the gerbil CA1 hippocampus. Hippocampus 1997; 7:511-23. [PMID: 9347348 DOI: 10.1002/(sici)1098-1063(1997)7:5<511::aid-hipo7>3.0.co;2-j] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Following transient cerebral ischemia, pyramidal cells within area CA1 of the hippocampus exhibit delayed neuronal death. While interneurons within this sector continue to survive long-term, there is evidence that some interneurons in area CA1 are vulnerable to damage. To determine the nature of vulnerability in a neurochemically heterogeneous population of interneurons throughout area CA1, we examined the labeling of gamma-aminobutyric acid (GABA)ergic interneurons with an antibody to the GABAA receptor alpha 1-subunit 1-35 days following cerebral ischemia in the Mongolian gerbil. Unlike some other GABA interneuron markers, this antibody labels both the dendrites and soma of interneurons, allowing dendritic structure to be examined. Three to four days following ischemia, the pyramidal cells in area CA1 had degenerated, and the alpha 1-subunit-positive interneurons in all layers of area CA1 had developed severely beaded dendrites. At longer survival times (21-35 days), the alpha 1-subunit-immunolabeled dendrites of these interneurons had a fragmented appearance. In contrast, interneurons bordering str. oriens and alveus typically exhibited normal dendritic morphology. Despite the pathologic changes, there was no evidence of interneuron loss in area CA1 up to 35 days post-ischemia. Normal interneuron morphology was also observed in area CA3 and dentate gyrus, regions where neither pyramidal neurons nor granule cells, respectively, die following 5 min of cerebral ischemia. To determine if the ischemia-induced changes in interneuron morphology could be prevented, diazepam was administered 30 and 90 min following ischemia. Diazepam produces long-term neuroprotection of area CA1 pyramidal neurons. In gerbils sacrificed 35 days after ischemia, diazepam markedly attenuated the dendritic beading of the area CA1 interneurons. In addition, the dendrites did not display the fragmented labeling by the alpha 1-subunit antibody. Thus, despite their long-term survival, CA1 hippocampal interneurons in the gerbil can express severe structural abnormalities after transient cerebral ischemia coincident with pyramidal cell degeneration, and the injury to the dendrites can be prevented by the neuroprotectant diazepam.
Collapse
Affiliation(s)
- J R Inglefield
- Department of Pharmacology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
43
|
Experimental neuronal protection in cerebral ischaemia Part II: Potential neuroprotective drugs. J Clin Neurosci 1997; 4:290-310. [DOI: 10.1016/s0967-5868(97)90096-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/1996] [Accepted: 06/04/1996] [Indexed: 01/01/2023]
|
44
|
Akins PT, Liu PK, Hsu CY. Immediate early gene expression in response to cerebral ischemia. Friend or foe? Stroke 1996; 27:1682-7. [PMID: 8784149 DOI: 10.1161/01.str.27.9.1682] [Citation(s) in RCA: 143] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Cerebral ischemia is a potent modulator of gene expression. Immediate early genes undergo rapid induction after both global and focal cerebral ischemia. Many immediate early genes code for transcription factors. Additional genes, including those encoding for neurotrophic factors and neurotransmitter systems, are induced in a delayed fashion after cerebral ischemia. The functional significance of early and late gene regulation after cerebral ischemia requires further investigation. These changes may be beneficial (friend) or detrimental (foe). Many of the genes are likely neuroprotective and important for recovery, but others may be involved in ischemic cell death mediated by apoptosis. SUMMARY OF REVIEW We review evidence that supports the hypothesis that cell death after cerebral ischemia occurs through the dual pathways of ischemic necrosis and apoptosis. CONCLUSIONS Gene regulation, including immediate early genes, is required for programmed neuronal death after trophic factor deprivation and is predicted to be involved in apoptosis triggered by cerebral ischemia. Novel therapies following cerebral ischemia may be directed at genes mediating either recovery or apoptosis.
Collapse
Affiliation(s)
- P T Akins
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | |
Collapse
|
45
|
Broussard DL, Li X, Altschuler SM. Localization of GABAA alpha 1 mRNA subunit in the brainstem nuclei controlling esophageal peristalsis. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1996; 40:143-7. [PMID: 8840023 DOI: 10.1016/0169-328x(96)00080-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The nucleus of the solitary tract, the site of esophageal premotor neurons (PMN), is tonically inhibited by GABAergic neurons via the GABAA receptor. We investigated the expression of GABAA alpha 1 subunit mRNA within esophageal PMNs of the NTS utilizing transynaptic tracing with pseudorabies virus and nonisotopic in-situ hybridization. Double-labeling studies revealed that the majority of PRV-immunoreactive cells also expressed GABAA alpha 1 mRNA. The expression of GABAA subunits supports a role for GABA in the brainstem circuit controlling esophageal peristalsis.
Collapse
Affiliation(s)
- D L Broussard
- Division of Gastrenterology and Nutrition, Children's Hospital of Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
46
|
Abstract
Cortical structures are often critically affected by ischemic and traumatic lesions which may cause transient or permanent functional disturbances. These disorders consist of changes in the membrane properties of single cells and alterations in synaptic network interactions within and between cortical areas including large-scale reorganizations in the representation of the peripheral input. Prominent functional modifications consisting of massive membrane depolarizations, suppression of intracortical inhibitory synaptic mechanisms and enhancement of excitatory synaptic transmission can be observed within a few minutes following the onset of cortical hypoxia or ischemia and probably represent the trigger signals for the induction of neuronal hyperexcitability, irreversible cellular dysfunction and cell death. Pharmacological manipulation of these early events may therefore be the most effective approach to control ischemia and lesion induced disturbances and to attenuate long-term neurological deficits. The complexity of secondary structural and functional alterations in cortical and subcortical structures demands an early and powerful intervention before neuronal damage expands to intact regions. The unsatisfactory clinical experience with calcium and N-methyl-D-aspartate antagonists suggests that this result might be achieved with compounds that show a broad spectrum of actions at different ligand-activated receptors, voltage-dependent channels and that also act at the vascular system. Whether the same therapy strategies developed for the treatment of ischemic injury in the adult brain may be applied for the immature cortex is questionable, since young cortical networks with a high degree of synaptic plasticity reveal a different response pattern to hypoxic and ischemic insults. Age-dependent molecular biological, morphological and physiological parameters contribute to an enhanced susceptibility of the immature brain to these noxae during early ontogenesis and have to be investigated in more detail for the development of adequate clinical therapy.
Collapse
Affiliation(s)
- H J Luhmann
- Department of Neurophysiology, University of Düsseldorf, Germany.
| |
Collapse
|
47
|
Small DL, Buchan AM. NMDA antagonists: their role in neuroprotection. INTERNATIONAL REVIEW OF NEUROBIOLOGY 1996; 40:137-71. [PMID: 8989620 DOI: 10.1016/s0074-7742(08)60719-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- D L Small
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Canada
| | | |
Collapse
|
48
|
Clément Y. Structural and pharmacological aspects of the GABAA receptor: involvement in behavioral pathogenesis. JOURNAL OF PHYSIOLOGY, PARIS 1996; 90:1-13. [PMID: 8803850 DOI: 10.1016/0928-4257(96)87164-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The gamma-aminobutyric acidA (GABAA) receptor is a complex hetero-oligomeric protein. It is composed of several subunits which assemble to form a functional chloride channel. The precise molecular organization of the receptor is as yet unknown. In the first part, we review recent literature dealing with the molecular and pharmacological aspects of the GABAA receptor, the second part will review some of the pathologies probably associated with gene defects and/or quantitative differential expression of transcripts encoding GABAA receptor subunits.
Collapse
Affiliation(s)
- Y Clément
- URA-CNRS 1957, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| |
Collapse
|
49
|
Fritschy JM, Mohler H. GABAA-receptor heterogeneity in the adult rat brain: differential regional and cellular distribution of seven major subunits. J Comp Neurol 1995; 359:154-94. [PMID: 8557845 DOI: 10.1002/cne.903590111] [Citation(s) in RCA: 969] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
GABAA-receptors display an extensive structural heterogeneity based on the differential assembly of a family of at least 15 subunits (alpha 1-6, beta 1-3, gamma 1-3, delta, rho 1-2) into distinct heteromeric receptor complexes. The subunit composition of receptor subtypes is expected to determine their physiological properties and pharmacological profiles, thereby contributing to flexibility in signal transduction and allosteric modulation. In heterologous expression systems, functional receptors require a combination of alpha-, beta-, and gamma-subunit variants, the gamma 2-subunit being essential to convey a classical benzodiazepine site to the receptor. The subunit composition and stoichiometry of native GABAA-receptor subtypes remain unknown. The aim of this study was to identify immunohistochemically the main subunit combinations expressed in the adult rat brain and to allocate them to identified neurons. The regional and cellular distribution of seven major subunits (alpha 1, alpha 2, alpha 3, alpha 5, beta 2,3, gamma 2, delta) was visualized by immunoperoxidase staining with subunit-specific antibodies (the beta 2- and beta 3-subunits were covisualized with the monoclonal antibody bd-17). Putative receptor subtypes were identified on the basis of colocalization of subunits within individual neurons, as analyzed by confocal laser microscopy in double- and triple-immunofluorescence staining experiments. The results reveal an extraordinary heterogeneity in the distribution of GABAA-receptor subunits, as evidenced by abrupt changes in immunoreactivity along well-defined cytoarchitectonic boundaries and by pronounced differences in the cellular distribution of subunits among various types of neurons. Thus, functionally and morphologically diverse neurons were characterized by a distinct GABAA-receptor subunit repertoire. The multiple staining experiments identified 12 subunit combinations in defined neurons. The most prevalent combination was the triplet alpha 1/beta 2,3/gamma 2, detected in numerous cell types throughout the brain. An additional subunit (alpha 2, alpha 3, or delta) sometimes was associated with this triplet, pointing to the existence of receptors containing four subunits. The triplets alpha 2/beta 2,3/gamma 2, alpha 3/beta 2,3/gamma 2, and alpha 5/beta 2,3/gamma 2 were also identified in discrete cell populations. The prevalence of these seven combinations suggest that they represent major GABAA-receptor subtypes. Five combinations also apparently lacked the beta 2,3-subunits, including one devoid of gamma 2-subunit (alpha 1/alpha 2/gamma 2, alpha 2/gamma 2, alpha 3/gamma 2, alpha 2/alpha 3/gamma 2, alpha 2/alpha 5/delta).(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- J M Fritschy
- Institute of Pharmacology, University of Zürich, Switzerland
| | | |
Collapse
|
50
|
Heurteaux C, Lauritzen I, Widmann C, Lazdunski M. Essential role of adenosine, adenosine A1 receptors, and ATP-sensitive K+ channels in cerebral ischemic preconditioning. Proc Natl Acad Sci U S A 1995; 92:4666-70. [PMID: 7753861 PMCID: PMC42005 DOI: 10.1073/pnas.92.10.4666] [Citation(s) in RCA: 449] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Preconditioning with sublethal ischemia protects against neuronal damage after subsequent lethal ischemic insults in hippocampal neurons. A pharmacological approach using agonists and antagonists at the adenosine A1 receptor as well as openers and blockers of ATP-sensitive K+ channels has been combined with an analysis of neuronal death and gene expression of subunits of glutamate and gamma-aminobutyric acid receptors, HSP70, c-fos, c-jun, and growth factors. It indicates that the mechanism of ischemic tolerance involves a cascade of events including liberation of adenosine, stimulation of adenosine A1 receptors, and, via these receptors, opening of sulfonylurea-sensitive ATP-sensitive K+ channels.
Collapse
Affiliation(s)
- C Heurteaux
- Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | | | | | | |
Collapse
|