1
|
Emmons-Bell M, Durant F, Tung A, Pietak A, Miller K, Kane A, Martyniuk CJ, Davidian D, Morokuma J, Levin M. Regenerative Adaptation to Electrochemical Perturbation in Planaria: A Molecular Analysis of Physiological Plasticity. iScience 2019; 22:147-165. [PMID: 31765995 PMCID: PMC6881696 DOI: 10.1016/j.isci.2019.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/01/2019] [Accepted: 11/05/2019] [Indexed: 12/29/2022] Open
Abstract
Anatomical homeostasis results from dynamic interactions between gene expression, physiology, and the external environment. Owing to its complexity, this cellular and organism-level phenotypic plasticity is still poorly understood. We establish planarian regeneration as a model for acquired tolerance to environments that alter endogenous physiology. Exposure to barium chloride (BaCl2) results in a rapid degeneration of anterior tissue in Dugesia japonica. Remarkably, continued exposure to fresh solution of BaCl2 results in regeneration of heads that are insensitive to BaCl2. RNA-seq revealed transcriptional changes in BaCl2-adapted heads that suggests a model of adaptation to excitotoxicity. Loss-of-function experiments confirmed several predictions: blockage of chloride and calcium channels allowed heads to survive initial BaCl2 exposure, inducing adaptation without prior exposure, whereas blockade of TRPM channels reversed adaptation. Such highly adaptive plasticity may represent an attractive target for biomedical strategies in a wide range of applications beyond its immediate relevance to excitotoxicity preconditioning. Exposure to BaCl2 causes the heads of Dugesia japonica to degenerate Prolonged exposure to BaCl2 results in regeneration of a BaCl2-insensitive head Ion channel expression is altered in the head to compensate for excitotoxic stress TRPMa is upregulated in BaCl2-treated animals; blocking TRPM prevents adaptation
Collapse
Affiliation(s)
- Maya Emmons-Bell
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Fallon Durant
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Angela Tung
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Alexis Pietak
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Kelsie Miller
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Anna Kane
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Devon Davidian
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Junji Morokuma
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA; Department of Biology, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
2
|
Walters DC, Jansen EEW, Ainslie GR, Salomons GS, Brown MN, Schmidt MA, Roullet J, Gibson KM. Preclinical tissue distribution and metabolic correlations of vigabatrin, an antiepileptic drug associated with potential use-limiting visual field defects. Pharmacol Res Perspect 2019; 7:e00456. [PMID: 30631446 PMCID: PMC6321982 DOI: 10.1002/prp2.456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 01/30/2023] Open
Abstract
Vigabatrin (VGB; (S)-(+)/(R)-(-) 4-aminohex-5-enoic acid), an antiepileptic irreversibly inactivating GABA transaminase (GABA-T), manifests use-limiting ocular toxicity. Hypothesizing that the active S enantiomer of VGB would preferentially accumulate in eye and visual cortex (VC) as one potential mechanism for ocular toxicity, we infused racemic VGB into mice via subcutaneous minipump at 35, 70, and 140 mg/kg/d (n = 6-8 animals/dose) for 12 days. VGB enantiomers, total GABA and β-alanine (BALA), 4-guanidinobutyrate (4-GBA), and creatine were quantified by mass spectrometry in eye, brain, liver, prefrontal cortex (PFC), and VC. Plasma VGB concentrations increased linearly by dose (3 ± 0.76 (35 mg/kg/d); 15.1 ± 1.4 (70 mg/kg/d); 34.6 ± 3.2 μmol/L (140 mg/kg/d); mean ± SEM) with an S/R ratio of 0.74 ± 0.02 (n = 14). Steady state S/R ratios (35, 70 mg/kg/d doses) were highest in eye (5.5 ± 0.2; P < 0.0001), followed by VC (3.9 ± 0.4), PFC (3.6 ± 0.3), liver (2.9 ± 0.1), and brain (1.5 ± 0.1; n = 13-14 each). Total VGB content of eye exceeded that of brain, PFC and VC at all doses. High-dose VGB diminished endogenous metabolite production, especially in PFC and VC. GABA significantly increased in all tissues (all doses) except brain; BALA increases were confined to liver and VC; and 4-GBA was prominently increased in brain, PFC and VC (and eye at high dose). Linear correlations between enantiomers and GABA were observed in all tissues, but only in PFC/VC for BALA, 4-GBA, and creatine. Preferential accumulation of the VGB S isomer in eye and VC may provide new insight into VGB ocular toxicity.
Collapse
Affiliation(s)
- Dana C. Walters
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| | - Erwin E. W. Jansen
- Metabolic LaboratoryDepartment of Clinical ChemistryAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Garrett R. Ainslie
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| | - Gajja S. Salomons
- Metabolic LaboratoryDepartment of Clinical ChemistryAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Madalyn N. Brown
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| | - Michelle A. Schmidt
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| | - Jean‐Baptiste Roullet
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| | - K. M. Gibson
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashington
| |
Collapse
|
3
|
Rempe RG, Hartz AMS, Bauer B. Matrix metalloproteinases in the brain and blood-brain barrier: Versatile breakers and makers. J Cereb Blood Flow Metab 2016; 36:1481-507. [PMID: 27323783 PMCID: PMC5012524 DOI: 10.1177/0271678x16655551] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/26/2016] [Indexed: 02/01/2023]
Abstract
Matrix metalloproteinases are versatile endopeptidases with many different functions in the body in health and disease. In the brain, matrix metalloproteinases are critical for tissue formation, neuronal network remodeling, and blood-brain barrier integrity. Many reviews have been published on matrix metalloproteinases before, most of which focus on the two best studied matrix metalloproteinases, the gelatinases MMP-2 and MMP-9, and their role in one or two diseases. In this review, we provide a broad overview of the role various matrix metalloproteinases play in brain disorders. We summarize and review current knowledge and understanding of matrix metalloproteinases in the brain and at the blood-brain barrier in neuroinflammation, multiple sclerosis, cerebral aneurysms, stroke, epilepsy, Alzheimer's disease, Parkinson's disease, and brain cancer. We discuss the detrimental effects matrix metalloproteinases can have in these conditions, contributing to blood-brain barrier leakage, neuroinflammation, neurotoxicity, demyelination, tumor angiogenesis, and cancer metastasis. We also discuss the beneficial role matrix metalloproteinases can play in neuroprotection and anti-inflammation. Finally, we address matrix metalloproteinases as potential therapeutic targets. Together, in this comprehensive review, we summarize current understanding and knowledge of matrix metalloproteinases in the brain and at the blood-brain barrier in brain disorders.
Collapse
Affiliation(s)
- Ralf G Rempe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
4
|
Llorente IL, Perez-Rodriguez D, Martínez-Villayandre B, Dos-Anjos S, Darlison MG, Poole AV, Fernández-López A. GABA(A) receptor chloride channels are involved in the neuroprotective role of GABA following oxygen and glucose deprivation in the rat cerebral cortex but not in the hippocampus. Brain Res 2013; 1533:141-51. [PMID: 23969196 DOI: 10.1016/j.brainres.2013.08.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/11/2013] [Accepted: 08/12/2013] [Indexed: 01/23/2023]
Abstract
Assays on "ex vivo" sections of rat hippocampus and rat cerebral cortex, subjected to oxygen and glucose deprivation (OGD) and a three-hour reperfusion-like (RL) recovery, were performed in the presence of either GABA or the GABA(A) receptor binding site antagonist, bicuculline. Lactate dehydrogenase (LDH) and propidium iodide were used to quantify cell mortality. We also measured, using real-time quantitative polymerase chain reaction (qPCR), the early transcriptional response of a number of genes of the glutamatergic and GABAergic systems. Specifically, glial pre- and post-synaptic glutamatergic transporters (namely GLAST1a, EAAC-1, GLT-1 and VGLUT1), three GABAA receptor subunits (α1, β2 and γ2), and the GABAergic presynaptic marker, glutamic acid decarboxylase (GAD65), were studied. Mortality assays revealed that GABAA receptor chloride channels play an important role in the neuroprotective effect of GABA in the cerebral cortex, but have a much smaller effect in the hippocampus. We also found that GABA reverses the OGD-dependent decrease in GABA(A) receptor transcript levels, as well as mRNA levels of the membrane and vesicular glutamate transporter genes. Based on the markers used, we conclude that OGD results in differential responses in the GABAergic presynaptic and postsynaptic systems.
Collapse
Affiliation(s)
- Irene L Llorente
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, 24071 León, Spain
| | | | | | | | | | | | | |
Collapse
|
5
|
Shoeb Ahmad S, Abdul Ghani S, Hemalata Rajagopal T. Current Concepts in the Biochemical Mechanisms of Glaucomatous Neurodegeneration. J Curr Glaucoma Pract 2013; 7:49-53. [PMID: 26997782 PMCID: PMC4741173 DOI: 10.5005/jp-journals-10008-1137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 10/19/2012] [Indexed: 12/31/2022] Open
Abstract
Glaucoma is now regarded as a neurodegenerative disorder. A number of theories including the mechanical and vascular models have been used to explain the pathogenesis of glaucoma. However, there is now increasing evidence of biochemical molecules which may play a part in it's causation. These biochemical mechanisms include the role of excitatory aminoacids, caspases, protein kinases, oxygen free radicals, nitric oxide, TNF-alpha, neurotrophins and metalloproteins. This paper reviews these new developments which form the biochemical basis of glaucomatous neural degeneration. How to cite this article: Ahmad SS, Ghani SA, Rajagopal TH. Current Concepts in the Biochemical Mechanisms of Glaucomatous Neurodegeneration. J Current Glau Prac 2013;7(2):49-53.
Collapse
Affiliation(s)
- Syed Shoeb Ahmad
- Ophthalmologist, Department of Ophthalmology, Queen Elizabeth, Hospital Kota Kinabalu, Malaysia
| | - Shuaibah Abdul Ghani
- Ophthalmologist, Department of Ophthalmology, Queen Elizabeth, Hospital Kota Kinabalu, Malaysia
| | | |
Collapse
|
6
|
Bae JS, Simon NG, Menon P, Vucic S, Kiernan MC. The puzzling case of hyperexcitability in amyotrophic lateral sclerosis. J Clin Neurol 2013; 9:65-74. [PMID: 23626643 PMCID: PMC3633193 DOI: 10.3988/jcn.2013.9.2.65] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 08/24/2012] [Accepted: 08/24/2012] [Indexed: 12/11/2022] Open
Abstract
The development of hyperexcitability in amyotrophic lateral sclerosis (ALS) is a well-known phenomenon. Despite controversy as to the underlying mechanisms, cortical hyperexcitability appears to be closely related to the interplay between excitatory corticomotoneurons and inhibitory interneurons. Hyperexcitability is not a static phenomenon but rather shows a pattern of progression in a spatiotemporal aspect. Cortical hyperexcitability may serve as a trigger to the development of anterior horn cell degeneration through a 'dying forward' process. Hyperexcitability appears to develop during the early disease stages and gradually disappears in the advanced stages of the disease, linked to the destruction of corticomotorneuronal pathways. As such, a more precise interpretation of these unique processes may provide new insight regarding the pathophysiology of ALS and its clinical features. Recently developed technologies such as threshold tracking transcranial magnetic stimulation and automated nerve excitability tests have provided some clues about underlying pathophysiological processes linked to hyperexcitability. Additionally, these novel techniques have enabled clinicians to use the specific finding of hyperexcitability as a useful diagnostic biomarker, enabling clarification of various ALS-mimic syndromes, and the prediction of disease development in pre-symptomatic carriers of familial ALS. In terms of nerve excitability tests for peripheral nerves, an increase in persistent Na+ conductances has been identified as a major determinant of peripheral hyperexcitability in ALS, inversely correlated with the survival in ALS. As such, the present Review will focus primarily on the puzzling theory of hyperexcitability in ALS and summarize clinical and pathophysiological implications for current and future ALS research.
Collapse
Affiliation(s)
- Jong Seok Bae
- Department of Neurology, College of Medicine, Inje University, Busan, Korea. ; Neuroscience Research Australia, Sydney, Australia
| | | | | | | | | |
Collapse
|
7
|
Heim MK, Gidal BE. Vigabatrin-associated retinal damage: potential biochemical mechanisms. Acta Neurol Scand 2012; 126:219-28. [PMID: 22632110 DOI: 10.1111/j.1600-0404.2012.01684.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2012] [Indexed: 12/13/2022]
Abstract
Vigabatrin (VGB), an irreversible inhibitor of gamma-aminobutyric acid (GABA) transaminase, is approved as adjunct treatment of refractory partial seizures as well as infantile spasms. Although VGB has been proven to be effective, its use is limited by the risk of retinopathy and associated peripheral visual field defects. This review describes and analyzes current literature related to potential pathophysiologic mechanisms underlying VGB-mediated cellular toxicity. Animal data suggest that GABA mediates neural excitotoxicity. The amino acid taurine is concentrated in retinal cells, and deficiency of this amino acid may be involved in VGB-mediated retinal degeneration and possible phototoxicity.
Collapse
Affiliation(s)
- M. K. Heim
- School of Pharmacy; University of Wisconsin - Madison; Madison; WI; USA
| | - B. E. Gidal
- School of Pharmacy & Department of Neurology; University of Wisconsin - Madison; Madison; WI; USA
| |
Collapse
|
8
|
Rout U, Abdul-Rahman OA, Dhossche DM. An immunological basis of hyperphagia driven by GABAergic dysfunction in Prader-Willi Syndrome. Med Hypotheses 2012; 78:462-4. [PMID: 22289342 DOI: 10.1016/j.mehy.2011.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 12/27/2011] [Indexed: 10/14/2022]
Abstract
Impaired immune function is increasingly seen as a core element of various neurological, psychiatric, and developmental disorders but has not yet been investigated in subjects with Prader-Willi Syndrome. We hypothesize that the emergence and the progression of PWS may be regulated by immune dysfunction involving auto-antibodies and miRNA driven by GABAergic dysfunction. Future research testing this hypothesis is discussed.
Collapse
Affiliation(s)
- Ujjwal Rout
- Department of Surgery, Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, MS 39216-4505, USA.
| | | | | |
Collapse
|
9
|
Cesetti T, Ciccolini F, Li Y. GABA Not Only a Neurotransmitter: Osmotic Regulation by GABA(A)R Signaling. Front Cell Neurosci 2012; 6:3. [PMID: 22319472 PMCID: PMC3268181 DOI: 10.3389/fncel.2012.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/10/2012] [Indexed: 12/05/2022] Open
Abstract
Mature macroglia and almost all neural progenitor types express γ-aminobutyric (GABA) A receptors (GABAARs), whose activation by ambient or synaptic GABA, leads to influx or efflux of chloride (Cl−) depending on its electro-chemical gradient (ECl). Since the flux of Cl− is indissolubly associated to that of osmotically obliged water, GABAARs regulate water movements by modulating ion gradients. In addition, since water movements also occur through specialized water channels and transporters, GABAAR signaling could affect the movement of water by regulating the function of the channels and transporters involved, thereby affecting not only the direction of the water fluxes but also their dynamics. We will here review recent observations indicating that in neural cells GABAAR-mediated osmotic regulation affects the cellular volume thereby activating multiple intracellular signaling mechanisms important for cell proliferation, maturation, and survival. In addition, we will discuss evidence that the osmotic regulation exerted by GABA may contribute to brain water homeostasis in physiological and in pathological conditions causing brain edema, in which the GABAergic transmission is often altered.
Collapse
Affiliation(s)
- Tiziana Cesetti
- Department of Physiology and Pathophysiology, Interdisciplinary Center for Neurosciences, University of Heidelberg Heidelberg, Germany
| | | | | |
Collapse
|
10
|
Qian H, Feng Y, He X, Yang Y, Sung JH, Xia Y. Effects of inhibitory amino acids on expression of GABAA Rα and glycine Rα1 in hypoxic rat cortical neurons during development. Brain Res 2011; 1425:1-12. [PMID: 22018691 DOI: 10.1016/j.brainres.2011.09.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 09/21/2011] [Accepted: 09/21/2011] [Indexed: 10/17/2022]
Abstract
Recent studies suggest that GABA and glycine are protective to mature but toxic to immature cortical neurons during prolonged hypoxia. Since the action of these inhibitory amino acids is mediated by GABA and glycine receptors, the expression of these receptors is a critical factor in determining neuronal response to GABA(A) and glycine in hypoxia. Therefore, we asked whether in rat cortical neurons, 1) hypoxia alters the expression of the GABA and glycine receptors; 2) inhibitory amino acids change the course of GABA and glycine receptor expression; and 3) there are any differences between the immature and mature neurons. In cultured rat cortical neurons from day 4 (four days in vitro or DIV 4) to day 20 (DIV 20), we observed that 1) GABA(A)Rα and GlyRα1 underwent differential changes in expression during the development in vitro; 2) hypoxia for 3 days decreased GABA(A)Rα and GlyRα1 density in the neurons in-between DIV 4 and DIV 20, but did not induce a major change in immature (DIV 4) and mature (DIV 20) neurons; 3) during normoxia GABA, glycine and taurine decreased GABA(A)Rα and GlyRα1 density in the immature neurons, but had a tendency to increase the density in the mature neurons, except for taurine; 4) under hypoxia, all these amino acids decreased GABA(A)Rα and GlyRα1 density in most groups of the immature neurons with a slight effect on the mature neurons; and 5) δ-opioid receptor activation with DADLE increased GABA(A)Rα and GlyRα1 density in both the immature and mature neurons under normoxia and in the mature neurons under hypoxic condition. These data suggest that inhibitory amino acids differentially regulate the expression of GABA(A) and glycine receptors in rat cortical neurons under normoxic and hypoxic conditions with major differences between the immature and mature neurons.
Collapse
Affiliation(s)
- Hong Qian
- Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | |
Collapse
|
11
|
Neurovascular interaction and the pathophysiology of diabetic retinopathy. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:693426. [PMID: 21747832 PMCID: PMC3124285 DOI: 10.1155/2011/693426] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/11/2011] [Accepted: 01/25/2011] [Indexed: 01/08/2023]
Abstract
Diabetic retinopathy (DR) is the most severe of the several ocular complications of diabetes, and in the United States it is the leading cause of blindness among adults 20 to 74 years of age. Despite recent advances in our understanding of the pathogenesis of DR, there is a pressing need to develop novel therapeutic treatments that are both safe and efficacious. In the present paper, we identify a key mechanism involved in the development of the disease, namely, the interaction between neuronal and vascular activities. Numerous pathological conditions in the CNS have been linked to abnormalities in the relationship between these systems. We suggest that a similar situation arises in the diabetic retina, and we propose a logical strategy aimed at therapeutic intervention.
Collapse
|
12
|
Brady JD, Mohr C, Rossi DJ. Vesicular GABA release delays the onset of the Purkinje cell terminal depolarization without affecting tissue swelling in cerebellar slices during simulated ischemia. Neuroscience 2010; 168:108-17. [PMID: 20226232 DOI: 10.1016/j.neuroscience.2010.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/02/2010] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Neurosteroids that can enhance GABA(A) receptor sensitivity protect cerebellar Purkinje cells against transient episodes of global brain ischemia, but little is known about how ischemia affects GABAergic transmission onto Purkinje cells. Here we use patch-clamp recording from Purkinje cells in acutely prepared slices of rat cerebellum to determine how ischemia affects GABAergic signaling to Purkinje cells. In voltage-clamped Purkinje cells, exposing slices to solutions designed to simulate brain ischemia caused an early, partial suppression of the frequency of spontaneous inhibitory post synaptic currents (sIPSCs), but after 5-8 min GABA accumulated in the extracellular space around Purkinje cells, generating a large (approximately 17 nS), sustained GABA(A) receptor-mediated conductance. The sustained GABA(A) conductance occurred in parallel with an even larger (approximately 117 nS) glutamate receptor-mediated conductance, but blocking GABA(A) receptors did not affect the timing or magnitude of the glutamate conductance, and blocking glutamate receptors did not affect the timing or magnitude of the GABA(A) conductance. Despite the lack of interaction between GABA and glutamate, blocking GABA(A) receptors significantly accelerated the onset of the Purkinje cell "ischemic" depolarization (ID), as assessed with current-clamp recordings from Purkinje cells or field potential recordings in the dendritic field of the Purkinje cells. The Purkinje cell ID occurred approximately 2 min prior to the sustained glutamate release under control conditions and a further 1-2 min earlier when GABA(A) receptors were blocked. Tissue swelling, as assessed by monitoring light transmittance through the slice, peaked just after the ID, prior to the sustained glutamate release, but was not affected by blocking GABA(A) receptors. These data indicate that ischemia induces the Purkinje cell ID and tissue swelling prior to the sustained glutamate release, and that blocking GABA(A) receptors accelerates the onset of the ID without affecting tissue swelling. Taken together these data may explain why Purkinje cells are one of the most ischemia sensitive neurons in the brain despite lacking NMDA receptors, and why neurosteroids that enhance GABA(A) receptor function protect Purkinje cells against transient episodes of global brain ischemia.
Collapse
Affiliation(s)
- J D Brady
- Department of Behavioral Neuroscience, Oregon Health and Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | | | | |
Collapse
|
13
|
Watanabe S, Kimura T, Suenaga K, Wada S, Tsuda K, Kasama S, Takaoka T, Kajiyama K, Takeda M, Yoshikawa H. Decreased chloride levels of cerebrospinal fluid in patients with amyotrophic lateral sclerosis. J Neurol Sci 2009; 285:146-8. [DOI: 10.1016/j.jns.2009.06.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 06/11/2009] [Accepted: 06/12/2009] [Indexed: 11/16/2022]
|
14
|
Carunchio I, Mollinari C, Pieri M, Merlo D, Zona C. GAB(A) receptors present higher affinity and modified subunit composition in spinal motor neurons from a genetic model of amyotrophic lateral sclerosis. Eur J Neurosci 2009; 28:1275-85. [PMID: 18973555 DOI: 10.1111/j.1460-9568.2008.06436.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis is a neurodegenerative disease characterized by the selective degeneration of motor neurons in the spinal cord, brainstem and cerebral cortex. In this study we have analysed the electrophysiological properties of GABA(A) receptors and GABA(A) alpha1 and alpha2 subunits expression in spinal motor neurons in culture obtained from a genetic model of ALS (G93A) and compared with transgenic wild type SOD1 (SOD1) and their corresponding non transgenic litter mates (Control). Although excitotoxic motor neuron death has been extensively studied in relation to Ca(2+)-dependent processes, strong evidence indicates that excitotoxic cell death is also remarkably dependent on Cl(-) ions and on GABA(A) receptor activation. In this study we have analysed the electrophysiological properties of GABA(A) receptors and the expression of GABA(A)alpha(1) and alpha(2) subunits in cultured motor neurons obtained from a genetic model of amyotrophic lateral sclerosis (G93A) and compared them with transgenic wild-type Cu,Zn superoxide dismutase and their corresponding non-transgenic littermates (Control). In all tested motor neurons, the application of gamma-aminobutyric acid (GABA) (0.5-100 mum) evoked an inward current that was reversibly blocked by bicuculline (100 mum), thus indicating that it was mediated by the activation of GABA(A) receptors. Our results indicate that the current density at high GABA concentrations is similar in control, Cu,Zn superoxide dismutase and G93A motor neurons. However, the dose-response curve significantly shifted toward lower concentration values in G93A motor neurons and the extent of desensitization also increased in these neurons. Finally, multiplex single-cell real-time polymerase chain reaction and immunofluorescence revealed that the amount of GABA(A)alpha(1) subunit was significantly increased in G93A motor neurons, whereas the levels of alpha(2) subunit were unchanged. These data show that the functionality and expression of GABA(A) receptors are altered in G93A motor neurons inducing a higher Cl(-) influx into the cell with a possible consequent neuronal excitotoxicity acceleration.
Collapse
Affiliation(s)
- Irene Carunchio
- Department of Neuroscience, University of Rome 'Tor Vergata', Via Montpellier 1, 00173 Rome, Italy
| | | | | | | | | |
Collapse
|
15
|
Traustason S, Eysteinsson T, Agnarsson BA, Stefánsson E. GABA agonists fail to protect the retina from ischemia-reperfusion injury. Exp Eye Res 2008; 88:361-6. [PMID: 19101544 DOI: 10.1016/j.exer.2008.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 06/17/2008] [Accepted: 07/08/2008] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to test the hypothesis that ischemia/reperfusion injury in the rat retina may be ameliorated by reducing retinal metabolism with either hypothermia or inhibitory GABA agonists. The intraocular pressure of each right eye in rats was raised to 130 mm Hg for 60 min with the left eye serving as normal control. The rats were divided into four groups in terms of drug and hypothermia treatment: (1) Untreated ischemia, (2) Hypothermia, (3) Baclofen/midazolam and (4) Baclofen/muscimol. Electroretinogram was recorded before ischemia and again after 10-day reperfusion. Histological analysis with H&E staining and cell counts was performed. Untreated ischemia/reperfusion resulted in severely reduced ERG responses. The ERG b-wave was reduced from 423+/-144 microV to 130+/-91 microV (mean+/-SD, n=5). With hypothermia the ERG b-wave was reduced from 499+/-80 microV to 237+/-111 microV (n=4). With combinations of baclofen and midazolam the ERG b-wave was reduced from 432+/-96 microV to 104+/-67 microV (n=7). In baclofen/muscimol treated eyes the ERG b-wave went from 426+/-101 microV to 148+/-118 microV (n=6). The histological tissue damage was severe in untreated ischemia and the baclofen/midazolam and baclofen/muscimol groups, but less severe in the hypothermia group. The GABA agonists do not provide any protection in our ischemia/reperfusion model. Our results are consistent with earlier reports that hypothermia may be helpful in ischemic conditions in the retina.
Collapse
Affiliation(s)
- Sindri Traustason
- Department of Ophthalmology and Physiology, National University Hospital, University of Iceland, Reykjavík, Iceland
| | | | | | | |
Collapse
|
16
|
Okumichi H, Mizukami M, Kiuchi Y, Kanamoto T. GABAA receptors are associated with retinal ganglion cell death induced by oxidative stress. Exp Eye Res 2008; 86:727-33. [DOI: 10.1016/j.exer.2008.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/23/2008] [Accepted: 01/27/2008] [Indexed: 01/10/2023]
|
17
|
Herden CJ, Pardo NE, Hajela RK, Yuan Y, Atchison WD. Differential Effects of Methylmercury on γ-Aminobutyric Acid Type A Receptor Currents in Rat Cerebellar Granule and Cerebral Cortical Neurons in Culture. J Pharmacol Exp Ther 2007; 324:517-28. [DOI: 10.1124/jpet.107.123976] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
18
|
Mongin AA. Disruption of ionic and cell volume homeostasis in cerebral ischemia: The perfect storm. ACTA ACUST UNITED AC 2007; 14:183-93. [PMID: 17961999 DOI: 10.1016/j.pathophys.2007.09.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The mechanisms of brain tissue damage in stroke are strongly linked to the phenomenon of excitotoxicity, which is defined as damage or death of neural cells due to excessive activation of receptors for the excitatory neurotransmitters glutamate and aspartate. Under physiological conditions, ionotropic glutamate receptors mediate the processes of excitatory neurotransmission and synaptic plasticity. In ischemia, sustained pathological release of glutamate from neurons and glial cells causes prolonged activation of these receptors, resulting in massive depolarization and cytoplasmic Ca(2+) overload. High cytoplasmic levels of Ca(2+) activate many degradative processes that, depending on the metabolic status, cause immediate or delayed death of neural cells. This traditional view has been expanded by a number of observations that implicate Cl(-) channels and several types of non-channel transporter proteins, such as the Na(+),K(+),2Cl(-) cotransporter, Na(+)/H(+) exchanger, and Na(+)/Ca(2+) exchanger, in the development of glutamate toxicity. Some of these ion transporters increase tissue damage by promoting pathological cell swelling and necrotic cell death, while others contribute to a long-term accumulation of cytoplasmic Ca(2+). This brief review is aimed at illustrating how the dysregulation of various ion transport processes combine in a 'perfect storm' that disrupts neural ionic homeostasis and culminates in the irreversible damage and death of neural cells. The clinical relevance of individual transporters as targets for therapeutic intervention in stroke is also briefly discussed.
Collapse
Affiliation(s)
- Alexander A Mongin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Avenue (MC-136), Albany, NY 12208, USA
| |
Collapse
|
19
|
Abstract
Excitotoxicity is associated with stroke, brain trauma, and a number of neurodegenerative disorders. In the brain, during excitotoxic insults, neurons undergo rapid swelling in both the soma and dendrites. Focal swellings along the dendrites called varicosities are considered to be a hallmark of acute excitotoxic neuronal injury. However, it is not clear what pathway is involved in the neuronal anion flux that leads to the formation and resolution of excitotoxic varicosities. Here, we assessed the roles of the volume-sensitive outwardly rectifying (VSOR) Cl- channel in excitotoxic responses in mouse cortical neurons. Whole-cell patch-clamp recordings revealed that the VSOR Cl- channel in cultured neurons was activated by NMDA exposure. Moreover, robust expression of this channel on varicosities was confirmed by on-cell and nystatin-perforated vesicle patch techniques. VSOR channel blockers, but not blockers of GABA(A) receptors and Cl- transporters, abolished not only varicosity resolution after sublethal excitotoxic stimulation but also necrotic death after sustained varicosity formation induced by prolonged NMDA exposure in cortical neurons. The present slice-patch experiments demonstrated, for the first time, expression of the VSOR Cl- channels in somatosensory pyramidal neurons. NMDA-induced necrotic neuronal death in slice preparations was largely suppressed by a blocker of the VSOR Cl- channel but not of the GABA(A) receptor. These results indicate that VSOR Cl- channels exert dual, reciprocal actions on neuronal excitotoxicity by serving as major anionic pathways both for varicosity recovery after washout of an excitotoxic stimulant and for persistent varicosity formation under prolonged excitotoxic insults leading to necrosis in cortical neurons.
Collapse
Affiliation(s)
- Hana Inoue
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Yasunobu Okada
- Department of Cell Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| |
Collapse
|
20
|
Morales I, Dopico JG, Sabate M, Gonzalez-Hernandez T, Rodriguez M. Substantia nigra osmoregulation: taurine and ATP involvement. Am J Physiol Cell Physiol 2007; 292:C1934-41. [PMID: 17215320 DOI: 10.1152/ajpcell.00593.2006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An extracellular nonsynaptic taurine pool of glial origin was recently reported in the substantia nigra (SN). There is previous evidence showing taurine as an inhibitory neurotransmitter in the SN, but the physiological role of this nonsynaptic pool of taurine has not been explored. By using microdialysis methods, we studied the action of local osmolarity on the nonsynaptic taurine pool in the SN of the rat. Hypoosmolar pulses (285-80 mosM) administered in the SN by the microdialysis probe increased extrasynaptic taurine in a dose-dependent way, a response that was counteracted by compensating osmolarity with choline. The opposite effect (taurine decrease) was observed when osmolarity was increased. Under basal conditions, the blockade of either the AMPA-kainate glutamate receptors with 6-cyano-7-nitroquinoxaline-2,3-dionine disodium or the purinergic receptors with pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid modified the taurine concentration, suggesting that both receptors modulate the extrasynaptic pool of taurine. In addition, these drugs decreased the taurine response to hypoosmolar pulses, suggesting roles for glutamatergic and purinergic receptors in the taurine response to osmolarity. The participation of purinergic receptors was also supported by the fact that ATP (which, under basal conditions, increased the extrasynaptic taurine in a dose-dependent way) administered in doses saturating purinergic receptors also decreased the taurine response to hypoosmolarity. Taken together, present data suggest osmoregulation as a role of the nonsynaptic taurine pool of the SN, a function that also involves glutamate and ATP and that could influence the nigral cell vulnerability in Parkinson's disease.
Collapse
Affiliation(s)
- Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain
| | | | | | | | | |
Collapse
|
21
|
Kiewert C, Kumar V, Hildmann O, Rueda M, Hartmann J, Naik RS, Klein J. Role of GABAergic antagonism in the neuroprotective effects of bilobalide. Brain Res 2006; 1128:70-8. [PMID: 17134681 PMCID: PMC1865101 DOI: 10.1016/j.brainres.2006.10.042] [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] [Received: 08/16/2006] [Revised: 10/07/2006] [Accepted: 10/20/2006] [Indexed: 10/23/2022]
Abstract
Bilobalide, a constituent of Ginkgo biloba, has neuroprotective properties. Its mechanism of action is unknown but it was recently found to block GABA(A) receptors. The goal of this study was to test the potential role of a GABAergic mechanism for the neuroprotective activity of bilobalide. In rat hippocampal slices exposed to NMDA, release of choline indicates breakdown of membrane phospholipids. NMDA-induced choline release was almost completely blocked in the presence of bilobalide (10 microM) and under low-chloride conditions. Bicuculline (100 microM), a competitive antagonist at GABA(A) receptors, reduced NMDA-induced choline release to a small extent (-23%). GABA (100 microM) partially antagonized the inhibitory action of bilobalide. Exposure of hippocampal slices to NMDA also caused edema formation as measured by increases of tissue water content. NMDA-induced edema formation was suppressed by bilobalide and by low-chloride conditions. Bicuculline exerted partial protection (by 30%) while GABA reduced bilobalide's effect by about one third. To investigate bilobalide's interaction with GABA(A) receptors directly, we measured binding of [(35)S]-TBPS to rat cortical membranes. TBPS binding was competitively inhibited by bilobalide in the low micromolar range (IC(50)=3.7 microM). As a functional test, we determined (36)chloride flux in rat corticohippocampal synaptoneurosomes. GABA (100 microM) significantly increased (36)chloride flux (+65%), and this increase was blocked by bilobalide, but with low potency (IC(50): 39 microM). We conclude that, while antagonism of GABA(A) receptors may contribute to bilobalide's neuroprotective effects, additional mechanisms must be postulated to fully explain bilobalide's actions.
Collapse
Affiliation(s)
- Cornelia Kiewert
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, 1300 Coulter Dr., Amarillo, TX 79106, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Kumar V, Naik RS, Hillert M, Klein J. Effects of chloride flux modulators in an in vitro model of brain edema formation. Brain Res 2006; 1122:222-9. [PMID: 17014830 PMCID: PMC1698554 DOI: 10.1016/j.brainres.2006.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 09/05/2006] [Accepted: 09/06/2006] [Indexed: 10/24/2022]
Abstract
Brain edema is a serious consequence of hemispheric stroke and traumatic brain injury and contributes significantly to patient mortality. In the present study, we measured water contents in hippocampal slices as an in vitro model of edema formation. Excitotoxic conditions induced by N-methyl-D-aspartate (NMDA, 300 microM), as well as ischemia induced by oxygen-glucose deprivation (OGD), caused cellular edema formation as indicated by an increase of slice water contents. In the presence of furosemide, an inhibitor of the Na,K,Cl-cotransporter, NMDA-induced edema were reduced by 64% while OGD-induced edema were unaffected. The same observation, i.e., reduction of excitotoxic edema formation but no effect on ischemia-induced edema, was made with chloride transport inhibitors such as DIDS and niflumic acid. Under ischemic conditions, modulation of GABAA receptors by bicuculline, a GABA antagonist, or by diazepam, a GABAergic agonist, did not significantly affect edema formation. Further experiments demonstrated that low chloride conditions prevented NMDA-induced, but not OGD-induced, water influx. Omission of calcium ions had no effect. Our results show that NMDA-induced edema formation is highly dependent on chloride influx as it was prevented by low-chloride conditions and by various compounds that interfere with chloride influx. In contrast, OGD-induced edema observed in brain slices was not affected by modulators of chloride fluxes. The results are discussed with reference to ionic changes occurring during tissue ischemia.
Collapse
Affiliation(s)
- Vikas Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, 1300 Coulter Dr, Amarillo, TX 79106, USA
| | | | | | | |
Collapse
|
23
|
Hernández TD. Post-Traumatic Neural Depression and Neurobehavioral Recovery after Brain Injury. J Neurotrauma 2006; 23:1211-21. [PMID: 16928179 DOI: 10.1089/neu.2006.23.1211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There are an estimated 2 million traumatic brain injuries (TBIs) each year in the United States, making the yearly incidence eight times greater than that of breast cancer and 34 times greater than HIV/AIDS. Still, it remains a "silent epidemic" because TBI results in persistent neurobehavioral impairment, without necessarily imparting a physical scar. The present review is a comparative analysis of TBI research, both basic and applied, outlining the evidence that at least one component of the brain's innate response to insult (e.g., post-traumatic neural depression) is sufficiently well understood to be the target of additional clinical studies and therapeutic strategy development.
Collapse
Affiliation(s)
- Theresa D Hernández
- Department of Psychology and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
24
|
Dopico JG, González-Hernández T, Pérez IM, García IG, Abril AM, Inchausti JO, Rodríguez Díaz M. Glycine release in the substantia nigra: Interaction with glutamate and GABA. Neuropharmacology 2006; 50:548-57. [PMID: 16337663 DOI: 10.1016/j.neuropharm.2005.10.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Revised: 09/28/2005] [Accepted: 10/25/2005] [Indexed: 01/25/2023]
Abstract
Previous studies have reported a high number of glycine (GLY) receptors in the substantia nigra (SN) but a low number of GLY-neurons, suggesting that taurine, a partial agonist of GLY-receptors, is the natural substrate for SN GLY-receptors. By using microdialysis to quantify amino acids in the extracellular space of the SN, we observed an extracellular pool of GLY in the rat that increased after depolarizing with high-K+ in a Ca2+-dependent manner and that diffuses through the extracellular space. GLY markedly increased after blocking either the tricarboxylic cycle with fluorocitrate or the glutamine synthetase activity with MSO. Because these products act selectively on glial cells, their effects show glia as a key cell in maintaining the extracellular pool of GLY in the SN. Extracellular GLY was modified by glutamate and glutamate receptor agonists. The local administration of GLY modified the extracellular concentration of GABA. Taken together, the complex regulation of the extracellular level of GLY, its possible glial origin and interaction with glutamate and GABA suggest a volume transmitter role for GLY in the SN, a possibility which also agrees with the recent finding of GLY-transporters in this centre.
Collapse
Affiliation(s)
- José García Dopico
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain
| | | | | | | | | | | | | |
Collapse
|
25
|
Pisani F, Costa C, Caccamo D, Mazzon E, Gorgone G, Oteri G, Calabresi P, Ientile R. Tiagabine and vigabatrin reduce the severity of NMDA-induced excitotoxicity in chick retina. Exp Brain Res 2006; 171:511-5. [PMID: 16418854 DOI: 10.1007/s00221-005-0298-1] [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] [Received: 03/29/2005] [Accepted: 10/31/2005] [Indexed: 10/25/2022]
Abstract
The possible neuroprotective effects of two GABAergic drugs, tiagabine (TGB) and vigabatrin (VGB), against N-methyl-D-aspartate (NMDA)-induced excitotoxicity have been investigated in the isolated chick embryo retina model. Retina segments were incubated either with NMDA alone (100 microM) or with NMDA and TGB or VGB (10-1,000 microM, added 5 min before NMDA). Retina damage was assessed after 24 h by measuring lactate dehydrogenase (LDH) activity present in the medium and by histological analysis. Both drugs reduced LDH release in a dose-dependent manner with comparable mean maximal values of 56.6-63.7% achieved at concentration of 1 mM. Histological analysis of retina slices was in line with the biochemical assays and showed partial preservation of drug exposed retina structure with reduced edema especially in the inner plexiform layer. The present data provide pharmacological evidence that both TGB and VGB reduce the severity of NMDA-induced excitotoxic damage. Although an increase in GABAergic transmission might play a role, this in itself is insufficient to explain the neuroprotective effect of the two drugs and the exact mechanism remains to be elucidated.
Collapse
Affiliation(s)
- Francesco Pisani
- Department of Neurosciences, Psychiatric and Anesthesiological Sciences, First Neurological Clinic, University of Messina, Messina, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
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: 1.5] [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
|
27
|
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: 3.5] [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
|
28
|
Babot Z, Cristòfol R, Suñol C. Excitotoxic death induced by released glutamate in depolarized primary cultures of mouse cerebellar granule cells is dependent on GABAAreceptors and niflumic acid-sensitive chloride channels. Eur J Neurosci 2005; 21:103-12. [PMID: 15654847 DOI: 10.1111/j.1460-9568.2004.03848.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Excitotoxic neuronal death has been linked to neurological and neurodegenerative diseases. Several studies have sought to clarify the involvement of Cl(-) channels in neuronal excitotoxicity using either N-methyl-D-aspartic acid (NMDA) or alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainic acid agonists. In this work we induced excitotoxic death in primary cultures of cerebellar granule cells by means of endogenously released glutamate. Excitotoxicity was provoked by exposure to high extracellular K(+) concentrations ([K(+)](o)) for 5 min. Under these conditions, a Ca(2+)-dependent release of glutamate was evoked. When extracellular glutamate concentration rose to between 2 and 4 microM, cell viability was significantly reduced by 30-40%. The NMDA receptor antagonists (MK-801 and D-2-amino-5-phosphonopentanoic acid) prevented cell death. Exposure to high [K(+)](o) produced a (36)Cl(-) influx which was significantly reduced by picrotoxinin. In addition, the GABA(A) receptor antagonists (bicuculline, picrotoxinin and SR 95531) protected cells from high [K(+)](o)-triggered excitotoxicity and reduced extracellular glutamate concentration. The Cl(-) channel blockers niflumic acid and 5-nitro-2-(3-phenylpropylamino)benzoic acid also exerted a neuroprotective effect and reduced extracellular glutamate concentration, even though they did not reduce high [K(+)](o)-induced (36)Cl(-) influx. Primary cultures of cerebellar granule cells also contain a population of GABAergic neurons that released GABA in response to high [K(+)](o). Chronic treatment of primary cultures with kainic acid abolished GABA release and rendered granule cells insensitive to high [K(+)](o) exposure, even though NMDA receptors were functional. Altogether, these results demonstrate that, under conditions of membrane depolarization, low micromolar concentrations of extracellular glutamate might induce an excitotoxic process through both NMDA and GABA(A) receptors and niflumic acid-sensitive Cl(-) channels.
Collapse
Affiliation(s)
- Zoila Babot
- Department of Neurochemistry, Institut d'Investigacions Biomediques de Barcelona, Consejo Superior de Investigaciones Cientificas, CSIC, IDIBAPS, Rossello 161, 08036 Barcelona, Spain
| | | | | |
Collapse
|
29
|
Allen NJ, Rossi DJ, Attwell D. Sequential release of GABA by exocytosis and reversed uptake leads to neuronal swelling in simulated ischemia of hippocampal slices. J Neurosci 2004; 24:3837-49. [PMID: 15084665 PMCID: PMC6729351 DOI: 10.1523/jneurosci.5539-03.2004] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
GABA release during cerebral energy deprivation (produced by anoxia or ischemia) has been suggested either to be neuroprotective, because GABA will hyperpolarize neurons and reduce release of excitotoxic glutamate, or to be neurotoxic, because activation of GABA(A) receptors facilitates Cl- entry into neurons and consequent cell swelling. We have used the GABA(A) receptors of hippocampal area CA1 pyramidal cells to sense the rise of [GABA](o) occurring in simulated ischemia. Ischemia evoked, after several minutes, a large depolarization to approximately -20 mV. Before this "anoxic depolarization," there was an increase in GABA release by exocytosis (spontaneous IPSCs). After the anoxic depolarization, there was a much larger, sustained release of GABA that was not affected by blocking action potentials, vesicular release, or the glial GABA transporter GAT-3 but was inhibited by blocking the neuronal GABA transporter GAT-1. Blocking GABA(A) receptors resulted in a more positive anoxic depolarization but decreased cell swelling at the time of the anoxic depolarization. The influence of GABA(A) receptors diminished in prolonged ischemia because glutamate release evoked by the anoxic depolarization inhibited GABA(A) receptor function by causing calcium entry through NMDA receptors. These data show that ischemia releases GABA initially by exocytosis and then by reversal of GAT-1 transporters and that the resulting Cl- influx through GABA(A) receptor channels causes potentially neurotoxic cell swelling.
Collapse
Affiliation(s)
- Nicola J Allen
- Department of Physiology, University College London, London WC1E 6BT, United Kingdom
| | | | | |
Collapse
|
30
|
Rodríguez Gil DJ, Carmona C, Negri G, Fiszer de Plazas S. Hypoxia differentially reduces GABA(A) receptor density during embryonic chick optic lobe development. Neurochem Res 2004; 29:681-6. [PMID: 15098929 DOI: 10.1023/b:nere.0000018838.43042.d4] [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: 11/12/2022]
Abstract
It has been demonstrated that the CNS is severely affected by hypoxic-ischemic insults during the prenatal-perinatal period, including imbalance in excitatory and inhibitory neurotransmitter release. Using a previously developed model of acute normobaric hypoxic hypoxia on chick embryos, we studied alterations observed both on [3H]GABA binding saturation parameters and on lactate concentration on successive embryonic days (ED). While maximal density of GABA binding sites (Bmax) from the low-affinity site was significantly reduced in an age-dependent manner, earlier stages of development (ED12 and 16) proving more vulnerable (ED12: control = 5.48 +/- 0.20, hypoxia = 3.90 +/- 0.39 pmol/mg prot, P < .05; ED16: control = 3.89 +/- 0.26, hypoxia = 2.80 +/- 0.28 pmol/mg prot, P < .05), ligand affinity (Kd) values and kinetic constants of the high-affinity site remained unaltered. Not unlikely, a physiological hypoxic state prevailing from ED17 up to hatching time rendered the whole embryo less sensitive to an externally induced hypoxic state (ED17: control = 2.93 +/- 0.06, hypoxia = 2.38 +/- 0.04 pmol/mg prot, P < .05; ED18: control = 2.97 +/- 0.12, hypoxia = 2.87 +/- 0.27 pmol/mg prot). Lactate levels in chick optic lobe homogenates were constant during development. The increase observed after hypoxic treatment compared to control value was significant at all stages studied, but increased percentage changes proved similar, indicating that all days of development equally perceive externally induced hypoxia. In conclusion, the present work demonstrates that after normobaric hypoxic hypoxia at different embryonic days, the embryo senses the externally induced hypoxic state as from ED12, but the GABA(A) receptor is differentially affected. It may be speculated that a different subunit composition of GABA(A) receptor is assembled in order to build a more stable receptor capable of resisting the physiological hypoxic state observed during the last few days before hatching.
Collapse
Affiliation(s)
- D J Rodríguez Gil
- Instituto de Biología Celular y Neurociencias, Prof. E. De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
31
|
Martikainen IK, Lauk K, Möykkynen T, Holopainen IE, Korpi ER, Uusi-Oukari M. Kainate down-regulates a subset of GABAA receptor subunits expressed in cultured mouse cerebellar granule cells. THE CEREBELLUM 2004; 3:27-38. [PMID: 15072265 DOI: 10.1080/14734220310020876] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The effect of kainate, an agonist selective for ionotropic AMPA/kainate type of glutamate receptors, on GABAA receptor subunit expression in cultured mouse cerebellar granule cells was studied using quantitative RT-PCR, ligand binding and electrophysiology. Chronic kainate treatment, without producing excitotoxicity, resulted in preferential, dose- and time-dependent down-regulation of alpha1, alpha6 and beta2 subunit mRNA expression, the expression of beta3, gamma2 and delta subunit mRNAs being less affected. The down-regulation was reversed by DNQX, an AMPA/kainate-selective glutamate receptor antagonist. A 14-day kainate treatment resulted in 46% decrease of total [3H]Ro 15-4513 binding to the benzodiazepine sites. Diazepam-insensitive [3H]Ro 15-4513 binding was decreased by 89% in accordance with very low amount of alpha6 subunit mRNA present. Diazepam-sensitive [3H]Ro 154513 binding was decreased only by 40%, contrasting >90% decrease in alpha1 subunit mRNA expression. However, this was consistent with lower potentiation of GABA-evoked currents in kainate-treated than control cells by the alpha1-selective benzodiazepine site ligand zolpidem, suggesting compensatory expression of alpha5 (and/or alpha2 or alpha3) subunits producing diazepam-sensitive but zolpidem-insensitive receptor subtypes. In conclusion, chronic kainate treatment of cerebellar granule cells selectively down-regulates oil, alpha6 and beta2 subunits resulting in altered GABAA receptor pharmacology.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cerebellum/cytology
- Cerebellum/drug effects
- Cerebellum/metabolism
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Down-Regulation/physiology
- GABA-A Receptor Antagonists
- Kainic Acid/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, GABA-A/biosynthesis
- Receptors, GABA-A/deficiency
- Receptors, GABA-A/genetics
- Receptors, Kainic Acid/agonists
- Receptors, Kainic Acid/genetics
- Receptors, Kainic Acid/metabolism
Collapse
Affiliation(s)
- Ilkka K Martikainen
- Department of Pharmacology and Clinical Pharmacology, University of Turku, Turku, Finland
| | | | | | | | | | | |
Collapse
|
32
|
Osborne NN, Casson RJ, Wood JPM, Chidlow G, Graham M, Melena J. Retinal ischemia: mechanisms of damage and potential therapeutic strategies. Prog Retin Eye Res 2004; 23:91-147. [PMID: 14766318 DOI: 10.1016/j.preteyeres.2003.12.001] [Citation(s) in RCA: 753] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Retinal ischemia is a common cause of visual impairment and blindness. At the cellular level, ischemic retinal injury consists of a self-reinforcing destructive cascade involving neuronal depolarisation, calcium influx and oxidative stress initiated by energy failure and increased glutamatergic stimulation. There is a cell-specific sensitivity to ischemic injury which may reflect variability in the balance of excitatory and inhibitory neurotransmitter receptors on a given cell. A number of animal models and analytical techniques have been used to study retinal ischemia, and an increasing number of treatments have been shown to interrupt the "ischemic cascade" and attenuate the detrimental effects of retinal ischemia. Thus far, however, success in the laboratory has not been translated to the clinic. Difficulties with the route of administration, dosage, and adverse effects may render certain experimental treatments clinically unusable. Furthermore, neuroprotection-based treatment strategies for stroke have so far been disappointing. However, compared to the brain, the retina exhibits a remarkable natural resistance to ischemic injury, which may reflect its peculiar metabolism and unique environment. Given the increasing understanding of the events involved in ischemic neuronal injury it is hoped that clinically effective treatments for retinal ischemia will soon be available.
Collapse
Affiliation(s)
- Neville N Osborne
- Nuffield Laboratory of Ophthalmology, University of Oxford, Walton Street, Oxford OX2 6AW, UK.
| | | | | | | | | | | |
Collapse
|
33
|
Petri S, Krampfl K, Hashemi F, Grothe C, Hori A, Dengler R, Bufler J. Distribution of GABAA receptor mRNA in the motor cortex of ALS patients. J Neuropathol Exp Neurol 2003; 62:1041-51. [PMID: 14575239 DOI: 10.1093/jnen/62.10.1041] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pathomechanism of amyotrophic lateral sclerosis (ALS) remains unclear. There is some evidence that excitotoxic cell death is involved in the degeneration of the motor nervous system, and that ligand-gated receptor channels play a role in the pathogenesis of the disease. Several electrophysiological and anatomical studies support the pathophysiological concept of an impaired inhibitory, namely GABAergic, control of the motoneurons in the cerebral cortex of ALS patients. The aim of our study was to investigate the expression of GABAA-receptor subunit mRNAs and the GABA synthesizing enzyme glutamic acid decarboxylase (GAD) in the motor cortex of ALS patients compared to tissue of control persons. We performed in situ hybridization histochemistry (ISH) on human postmortem motor cortex sections of ALS patients (n = 5) and age matched controls with no history of neurological disease (n = 5). The most intriguing finding was a significantly reduced mRNA expression of the alpha1-subunit in ALS patients while the level of beta1-subunit mRNA was elevated in the patients group. This may indicate specific alterations of the GABAA receptor subunit composition and result in distinct physiological and pharmacological properties of these receptors in ALS patients.
Collapse
Affiliation(s)
- Susanne Petri
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30623 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Andrés N, Malpesa Y, Rodríguez MJ, Mahy N. Low sensitivity of retina to AMPA-induced calcification. J Neurosci Res 2003; 72:543-8. [PMID: 12704816 DOI: 10.1002/jnr.10601] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Glutamate is involved in most CNS neurodegenerative diseases. In particular, retinal diseases such as retinal ischemia, retinitis pigmentosa, and diabetic retinopathy are associated with an excessive synaptic concentration of this neurotransmitter. To gain more insight into retinal excitotoxicity, we carried out a dose-response study in adult rats using alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA), a glutamate analogue. AMPA intraocular injections (between 0.27 and 10.8 nmol) caused no morphologic modification, but a 10.8 + 21 nmol double injection in a 10-day interval produced a lesion characterized by discrete neuronal loss, astroglial and microglial reactions, and calcium precipitation. Abundant calcium deposits similar to those present in rat and human brain excitotoxicity or hypoxia-ischemia neurodegeneration were detected by alizarin red staining within the retinal surface and the optic nerve. Glial reactivity, associated normally with astrocytes in the nerve fiber, was assessed in Müller cells. GABA immunoreactivity was detected not only in neuronal elements but also in Müller cells. In contrast to the high vulnerability of the brain to excitotoxin microinjection, AMPA-induced retinal neurodegeneration may provide a useful model of low central nervous system sensitivity to excitotoxicity.
Collapse
Affiliation(s)
- Noemí Andrés
- Unitat de Bioquímica, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
36
|
Rodríguez Gil DJ, Mitridate de Novara A, Fiszer de Plazas S. Acute hypoxic hypoxia alters GABA(A) receptor modulation by allopregnanolone and pentobarbital in embryonic chick optic lobe. Brain Res 2002; 954:294-9. [PMID: 12414112 DOI: 10.1016/s0006-8993(02)03357-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Using a previously developed model of acute normobaric hypoxic hypoxia on chick embryos, here we studied at embryonic day 12 the in vitro effect of two positive allosteric modulators of GABA binding, the barbiturate sodium pentobarbital and the neurosteroid allopregnanolone. In both cases an increase in E(max) values in membranes obtained from hypoxic embryos was observed. Studies of GABA-gated chloride influx showed that there were no differences in maximal chloride uptake between hypoxic and control membranes. We have already demonstrated that maximal density of GABA binding sites was decreased after hypoxia, suggesting that each of the remaining GABA(A) receptors display a greater chloride flux than controls. To further characterize GABA(A) receptor alterations, GABA-gated chloride influx modulated by the above barbiturate and neurosteroid was determined, finding that E(max) values were increased 60% and 42%, respectively. The increase in Cl(-) influx per receptor subsequent to hypoxic trauma, and the enhancement in the modulatory properties studied, may mediate neuronal damage by potential changes in subunit interaction at the GABA(A) receptor level.
Collapse
Affiliation(s)
- D J Rodríguez Gil
- Instituto de Biología Celular y Neurociencias, Professor E De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 1121 Buenos Aires, Argentina
| | | | | |
Collapse
|
37
|
Fåhraeus C, Theander S, Edman A, Grampp W. The K-Cl cotransporter in the lobster stretch receptor neurone--a kinetic analysis. J Theor Biol 2002; 217:287-309. [PMID: 12270275 DOI: 10.1006/jtbi.2002.3038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Experiments were performed to define quantitatively the substrate (K(+) and Cl(-)) dependence of the transport function (production of equally large and oppositely directed K(+)and Cl(-) flows/currents) of an earlier (Theander et al., 1999) identified electroneutral K-Cl cotransporter in the slowly adapting stretch receptor neurone of the European lobster. The experiments were based on microelectrode techniques. This allowed us to perform steady-state measurements of the so-called "instantaneous" current-voltage relationships (around a holding voltage of -65 mV after a blockage of the cell's action potential and hyperpolarization-activated currents) and intracellular ion concentrations at various settings of the extracellular K(+) and Cl(-) concentrations. From the results, we could then define steady-state values of all of the cell's non-KCl cotransporter K(+) and Cl(-) currents. Finally, the negative sums of the inferred non-KCl cotransporter K(+) and Cl(-) currents could be taken as equivalents of the K-Cl cotransporter's K(+) and Cl(-) currents for the reason that, in steady state, all membrane currents add up to zero. For the cotransporter currents, thus inferred for a range from 2.5/410.5 to 40.0/448.0 mM external K(+)/Cl(-), we found that their absolute values increased in a nonlinear fashion from about 5 nA cell(-1) at the lowest, to about 20 nA cell(-1) at the highest external K(+)/Cl(-) concentrations. Formally, this relationship could be reproduced by a Hill function-based enzyme kinetic expression simulating inward and outward transmembrane electroneutral ion transports. Following insertion of this expression into a comprehensive model of electrical membrane functions and intracellular solute and solvent control in the lobster stretch receptor neurone, the model predictions suggested that the K-Cl cotransporter does play an important role in (a) keeping intracellular Cl(-) low for a proper function of the cell's inhibitory system, and (b) enabling rapid transmembrane K(+) shifts that provide for a stabilization of the cell's membrane voltage and membrane excitability in cases of varying extracellular K(+) concentrations. The model predictions gave, however, no clear evidence that the K-Cl cotransporter is critically involved in the cell's volume regulation in conditions of varying extracellular osmolalities.
Collapse
Affiliation(s)
- C Fåhraeus
- Department of Physiological Sciences, Section of Neurophysiology, University of Lund, BMC F11 S-221 84, Lund, Sweden
| | | | | | | |
Collapse
|
38
|
Maus M, Glowinski J, Premont J. GABA is toxic for mouse striatal neurones through a transporter-mediated process. J Neurochem 2002; 82:763-73. [PMID: 12358781 DOI: 10.1046/j.1471-4159.2002.01011.x] [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: 11/20/2022]
Abstract
In the present study, GABA was shown to induce a necrotic neuronal death in cultured striatal neurones from mouse embryos. This effect did not depend on the activation of GABA(A), GABA(B) or GABA(C) receptors as it was neither antagonized by bicuculline, saclofen or picrotoxin, respectively, nor reproduced by the GABA receptor agonists, muscimol and baclofen. Excluding the participation of glutamate, GABA neurotoxicity persisted in the presence of either the antagonists of ionotropic and metabotropic glutamate receptors or glutamate pyruvate transaminase, which induces an immediate catabolism of glutamate. A GABA transport-associated process is involved in GABA neurotoxicity as nipecotic acid and NO 711, two inhibitors of the high-affinity neuronal GABA transporters (GAT-1, in particular), completely prevented the neurotoxic effect of GABA. The activation of a subset of G proteins is also implicated in the GABA transport-mediated neuronal death as GABA neurotoxicity was completely suppressed when striatal neurones were pre-treated with pertussis toxin. Further demonstrating the specificity of this neurotoxic process, GABA-induced neurotoxicity was not observed in cortical neurones which, in contrast to striatal neurones, are largely represented by glutamatergic neurones. In conclusion, our study suggests that glutamate is not the sole neurotransmitter that can be responsible for brain damage and that GABA neurotoxicity involves both GABA transport and G protein transduction pathways.
Collapse
Affiliation(s)
- Marion Maus
- INSERM U114, College de France, Paris, France
| | | | | |
Collapse
|
39
|
Zeevalk GD, Manzino L, Sonsalla PK. Protection of malonate-induced GABA but not dopamine loss by GABA transporter blockade in rat striatum. Exp Neurol 2002; 176:193-202. [PMID: 12093096 DOI: 10.1006/exnr.2002.7917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous work has shown that overstimulation of GABA(A) receptors can potentiate neuronal cell damage during excitotoxic or metabolic stress in vitro and that GABA(A) antagonists or GABA transport blockers are neuroprotective under these situations. Malonate, a reversible succinate dehydrogenase/mitochondrial complex II inhibitor, is frequently used in animals to model cell loss in neurodegenerative diseases such as Parkinson's and Huntington's diseases. To determine if GABA transporter blockade during mitochondrial impairment can protect neurons in vivo as compared with in vitro studies, rats received a stereotaxic infusion of malonate (2 micromol) into the left striatum to induce a metabolic stress. The nonsubstrate GABA transport blocker, NO711 (20 nmol) was infused in some rats 30 min before and 3 h following malonate infusion. After 1 week, dopamine and GABA levels in the striata were measured. Malonate caused a significant loss of striatal dopamine and GABA. Blockade of the GABA transporter significantly attenuated GABA, but not dopamine loss. In contrast with several in vitro reports, GABA(A) receptors were not a downstream mediator of protection by NO711. Intrastriatal infusion of malonate (2 micromol) plus or minus the GABA(A) receptor agonist muscimol (1 micromol), the GABA(A) Cl- binding site antagonist picrotoxin (50 nmol) or the GABA(B) receptor antagonist saclofen (33 nmol) did not modify loss of striatal dopamine or GABA when examined 1 week following infusion. These data show that GABA transporter blockade during mitochondrial impairment in the striatum provides protection to GABAergic neurons. GABA transporter blockade, which is currently a pharmacological strategy for the treatment of epilepsy, may thus also be beneficial in the treatment of acute and chronic conditions involving energy inhibition such as stroke/ischemia or Huntington's disease. These findings also point to fundamental differences between immature and adult neurons in the downstream involvement of GABA receptors during metabolic insult.
Collapse
Affiliation(s)
- Gail D Zeevalk
- Department of Neurology, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
40
|
Disease-specific human glycine receptor alpha1 subunit causes hyperekplexia phenotype and impaired glycine- and GABA(A)-receptor transmission in transgenic mice. J Neurosci 2002. [PMID: 11923415 DOI: 10.1523/jneurosci.22-07-02505.2002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hereditary hyperekplexia is caused by disinhibition of motoneurons resulting from mutations in the ionotropic receptor for the inhibitory neurotransmitter glycine (GlyR). To study the pathomechanisms involved in vivo, we generated and analyzed transgenic mice expressing the hyperekplexia-specific dominant mutant human GlyR alpha1 subunit 271Q. Tg271Q transgenic mice, in contrast to transgenic animals expressing a wild-type human alpha1 subunit (tg271R), display a dramatic phenotype similar to spontaneous and engineered mouse mutations expressing reduced levels of GlyR. Electrophysiological analysis in the ventral horn of the spinal cord of tg271Q mice revealed a diminished GlyR transmission. Intriguingly, an even larger reduction was found for GABA(A)-receptor-mediated inhibitory transmission, indicating that the expression of this disease gene not only affects the glycinergic system but also leads to a drastic downregulation of the entire postsynaptic inhibition. Therefore, the transgenic mice generated here provide a new animal model of systemic receptor interaction to study inherited and acquired neuromotor deficiencies at different functional levels and to develop novel therapeutic concepts for these diseases.
Collapse
|
41
|
Comaish IF, Gorman C, Brimlow GM, Barber C, Orr GM, Galloway NR. The effects of vigabatrin on electrophysiology and visual fields in epileptics: a controlled study with a discussion of possible mechanisms. Doc Ophthalmol 2002; 104:195-212. [PMID: 11999627 DOI: 10.1023/a:1014603229383] [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: 11/12/2022]
Abstract
PURPOSE To compare the visual electrophysiology and visual fields of patients taking vigabatrin to those of a control group of epileptics on other anti-epileptic drugs (AEDs). METHODS Fourteen epileptics treated with vigabatrin and 10 control patients treated with other AEDs underwent ERG and EOG. Goldmann visual fields were performed and analysed using standard software to measure areas contained within I4e isopters. RESULTS The cone and rod b-waves of the ERG, the oscillatory potential amplitudes and Arden indices were reduced in vigabatrin-treated subjects and the oscillatory potentials delayed. The Arden indices were reduced due to an increased dark trough. The areas contained within the I4e isopter of vigabatrin treated subjects were reduced compared to the control group and these areas correlated well with oscillatory potential amplitudes and b-wave amplitudes in the vigabatrin group only. CONCLUSIONS The use of vigabatrin is associated with a reduction of the ERG cone b-wave amplitude and oscillatory potentials which correlates with visual field loss. The Arden ratio is reduced in subjects taking vigabatrin but may recover after cessation. However, visual loss may persist in the presence of a recovered EOG. These findings suggest further effects of the drug than those mediated by GABA receptors, and support the contention that the cause of the field loss may be at least in part due to retinal effects. Possible mechanisms are discussed.
Collapse
Affiliation(s)
- I F Comaish
- Department of Ophthalmology, Queen's Medical Centre, Nottingham, UK.
| | | | | | | | | | | |
Collapse
|
42
|
Shen DW, Higgs MH, Salvay D, Olney JW, Lukasiewicz PD, Romano C. Morphological and electrophysiological evidence for an ionotropic GABA receptor of novel pharmacology. J Neurophysiol 2002; 87:250-6. [PMID: 11784747 DOI: 10.1152/jn.00620.2001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Evidence from toxicological studies suggested that an ionotropic GABA receptor of novel pharmacology (picrotoxin-insensitive, bicuculline-sensitive) exists in the chick embryo retina. In this report, we provide direct morphological and electrophysiological evidence for the existence of such an iGABA receptor. Chick embryo retinas (14-16 days old) incubated in the presence of kainic acid showed pronounced histopathology in all retinal layers. Maximal protection from this toxicity required a combination of bicuculline and picrotoxin. Individual application of the antagonists indicated that a picrotoxin-insensitive, bicuculline-sensitive GABA receptor is likely to be present on ganglion and amacrine, but not bipolar, cells. GABA currents in embryonic and mature chicken retinal neurons were measured by whole cell patch clamp. GABA was puffed at the dendritic processes in the IPL. Picrotoxin (500 microM, in the bath) eliminated all (>95%) the GABA current in the majority of ganglion and amacrine cells tested, but many cells possessed a substantial picrotoxin-insensitive component. This current was eliminated by bicuculline (200 microM). This current was not a transporter-associated current, since it was not altered by GABA transport blockers or sodium removal. The current-voltage relation was linear and reversed near E(Cl), as expected for a ligand-gated chloride current. Both pentobarbital and lorazepam enhanced the picrotoxin-insensitive current. We conclude that chicken retinal ganglion and amacrine cells express a GABA receptor that is GABA-A-like, in that it can be blocked by bicuculline, and positively modulated by barbiturates and benzodiazepines, but is insensitive to the noncompetitive blocker picrotoxin. Understanding the molecular properties of this receptor will be important for understanding both physiological GABA neurotransmission and the pathology of GABA receptor overactivation.
Collapse
Affiliation(s)
- D-W Shen
- Department of Ophthalmology and Visual Science, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
43
|
Chang Y, Xie Y, Weiss DS. Positive allosteric modulation by ultraviolet irradiation on GABA(A), but not GABA(C), receptors expressed in Xenopus oocytes. J Physiol 2001; 536:471-8. [PMID: 11600682 PMCID: PMC2278889 DOI: 10.1111/j.1469-7793.2001.0471c.xd] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
1. Recombinant rat GABA(A) (alpha1beta2, alpha1beta2gamma2, beta2gamma2) and human GABA(C) (rho1) receptors were expressed in Xenopus oocytes to examine the effect of ultraviolet (UV) light on receptor function. 2. GABA-induced currents in individual oocytes expressing GABA receptors were tested by two-electrode voltage clamp before, and immediately after, 312 nm UV irradiation. 3. UV irradiation significantly potentiated 10 microM GABA-induced currents in alpha1beta2gamma2 GABA receptors. The modulation was irradiation dose dependent, with a maximum potentiation of more than 3-fold. 4. The potentiation was partially reversible and decayed exponentially with a time constant of 8.2 +/- 1.2 min toward a steady-state level which was still significantly elevated (2.7 +/- 0.3-fold) compared to the control level. 5. The effect of UV irradiation on GABA(A) receptors varied with receptor subunit composition. UV irradiation decreased the EC50 of the alpha1beta2, alpha1beta2gamma2 and beta2gamma2 GABA(A) receptors, but exhibited no significant effect on the rho1 GABA(C) receptor. 6. UV irradiation also significantly increased the maximum current 2-fold in alpha1beta2 GABA(A) receptors with little effect on the maximum of alpha1beta2gamma2 (1.1-fold) or beta2gamma2 (1.1-fold) GABA(A) receptors. 7. The effect of UV irradiation on GABA(A) receptors did not overlap the effect of the GABA receptor- allosteric modulator, diazepam. 8. The UV effect on GABA(A) receptors was not prevented by the treatment of the oocytes before and during UV irradiation with one of the following free-radical scavengers: 40 mM D-mannitol, 40 mM imidazole or 40 mM sodium azide. In addition, the effect was not mimicked by the free-radical generator, H2O2. 9. Potential significance and mechanism(s) of the UV effect on GABA receptors are discussed.
Collapse
Affiliation(s)
- Y Chang
- Department of Neurobiology, University of Alabama at Birmingham, 1719 Sixth Avenue South, CIRC410, Birmingham, AL 35294-0021, USA
| | | | | |
Collapse
|
44
|
Gulyás AI, Sík A, Payne JA, Kaila K, Freund TF. The KCl cotransporter, KCC2, is highly expressed in the vicinity of excitatory synapses in the rat hippocampus. Eur J Neurosci 2001; 13:2205-17. [PMID: 11454023 DOI: 10.1046/j.0953-816x.2001.01600.x] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Immunocytochemical visualization of the neuron-specific K+/Cl- cotransporter, KCC2, at the cellular and subcellular level revealed an area- and layer-specific diffuse labelling, and a discrete staining outlining the somata and dendrites of some interneurons in all areas of the rat hippocampus. KCC2 was highly expressed in parvalbumin-containing interneurons, as well as in subsets of calbindin, calretinin and metabotropic glutamate receptor 1a-immunoreactive interneurons. During the first 2 postnatal weeks, an increase of KCC2 staining was observed in the molecular layer of the dentate gyrus, correlating temporally with the arrival of entorhinal cortical inputs. Subcellular localization demonstrated KCC2 in the plasma membranes. Immunoreactivity in principal cells was responsible for the diffuse staining found in the neuropil. In these cells, KCC2 was detected primarily in dendritic spine heads, at the origin of spines and, at a much lower level on the somata and dendritic shafts. KCC2 expression was considerably higher in the somata and dendrites of interneurons, most notably of parvalbumin-containing cells, as well as in the thorny excrescences of CA3 pyramidal cells and in the spines of spiny hilar and stratum lucidum interneurons. The data indicate that KCC2 is highly expressed in the vicinity of excitatory inputs in the hippocampus, perhaps in close association with extrasynaptic GABAA receptors. A high level of excitation is known to lead to a simultaneous net influx of Na+ and Cl-, as evidenced by dendritic swelling. KCC2 located in the same microenvironment may provide a Cl- extrusion mechanism to deal with both ion and water homeostasis in addition to its role in setting the driving force of Cl- currents involved in fast postsynaptic inhibition.
Collapse
Affiliation(s)
- A I Gulyás
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, P.O. Box 67, H-1450, Hungary
| | | | | | | | | |
Collapse
|
45
|
Stokes AH, Bernard LP, Nicklas WJ, Zeevalk GD. Attenuation of malonate toxicity in primary mesencephalic cultures using the GABA transport blocker, NO-711. J Neurosci Res 2001; 64:43-52. [PMID: 11276050 DOI: 10.1002/jnr.1052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cultured rat mesencephalic neurons were used to assess the effects of gamma-aminobutyric acid (GABA) transport blockers on toxicity caused by malonate, a reversible, competitive inhibitor of succinate dehydrogenase. Previous studies utilizing an ex vivo chick retinal preparation have shown that GABA release and cell swelling are early consequences of acute energy impairment and that GABA transport blockers attenuate this toxicity. The present results demonstrate that the nonsubstrate GABA transport blocker, NO-711 (1 nM-1 microM), dose-dependently protected cultured mesencephalic dopamine (DA) and GABA neurons from malonate-induced toxicity. Similar protection was demonstrated with nipecotic acid (1 mM) and SKF89976A (100 nM), substrate and nonsubstrate GABA transport blockers, respectively. These compounds by themselves produced no signs of toxicity, although nipecotic acid caused a long-term decrease in GABA uptake not associated with toxicity. Compounds which decrease intracellular reactive oxygen species (ROS) are protective in this model, but NO-711 did not prevent the rise in intracellular ROS induced by malonate, indicating its protective effects were downstream of ROS production. Supplementation of malonate treated cultures with the GABA(A) agonist, muscimol (10 microM), increased the toxicity toward the DA and GABA neuron populations. Antagonists at the GABA(A) and glycine receptors provided partial protection to both the GABA and DA neurons. These findings suggest that the GABA transporter, GABA(A), and/or glycine channels contribute to cell damage associated with energy impairment in this model.
Collapse
Affiliation(s)
- A H Stokes
- Department of Neurology, Robert Wood Johnson Medical School-UMDNJ, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
46
|
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
|
47
|
Johnston JB, Zhang K, Silva C, Shalinsky DR, Conant K, Ni W, Corbett D, Yong VW, Power C. HIV-1 Tat neurotoxicity is prevented by matrix metalloproteinase inhibitors. Ann Neurol 2001; 49:230-41. [PMID: 11220743 DOI: 10.1002/1531-8249(20010201)49:2<230::aid-ana43>3.0.co;2-o] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The release of potentially neurotoxic molecules by HIV-infected brain macrophages is accompanied by neuronal injury and death that results in the development of HIV-associated dementia (HAD). Among the potential neurotoxins implicated in the development of HAD is the HIV-1 transactivating protein, Tat. To investigate the mechanism by which Tat causes neurotoxicity, brain-derived Tat sequences from nondemented (Tat-ND) and demented (Tat-HAD) AIDS patients, which differed primarily in the augmenting region of Tat, were expressed in U937 monoblastoid cells and primary human macrophages. Cells expressing Tat-HAD protein exhibited elevated matrix metalloproteinase (MMP)-2 and -7 release and activation, but cells expressing Tat-ND did not exhibit enhanced MMP expression. Conditioned media from Tat-HAD-transfected cells caused significantly greater neuronal death (15.4 +/- 4.3%) than did Tat-ND (4.4 +/- 2.1%) or nontransfected (2.1 +/- 0.8%) cell-derived conditioned media. The neurotoxicity induced by Tat-HAD was inhibited by anti-MMP-2 or -7 antibodies (p < 0.005) but not by antibodies against MMP-9 or Tat. Similarly, scid/nod mice receiving striatal implants of Tat-HAD-transfected cells exhibited greater neurobehavioral abnormalities and neuronal loss (p < 0.005) than did animals receiving Tat-ND or nontransfected cells, which were reduced by treatment with the MMP inhibitor prinomastat (p < 0.005). These findings indicate that Tat causes neuronal death through an indirect mechanism that is Tat sequence dependent and involves the induction of MMPs.
Collapse
Affiliation(s)
- J B Johnston
- Department of Clinical Neuroscience, University of Calgary, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|