1
|
Zaczek R, Traynelis SF, Dingledine R, Koszalka GW, Laskowitz DT. Phase 1 Clinical Results for NP10679, a pH-sensitive GluN2B-selective N-methyl-d-aspartate Receptor Inhibitor. Clin Pharmacol Drug Dev 2023; 12:706-717. [PMID: 36642931 PMCID: PMC10329973 DOI: 10.1002/cpdd.1217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023]
Abstract
NP10679 is a context-dependent and subunit-selective negative allosteric modulator of N-methyl-d-aspartate (NMDA) receptors. It is a more potent inhibitor of GluN2B-containing NMDA receptors at the acidic levels of extracellular pH (eg, 6.9) found in the penumbral regions associated with cerebral ischemia than at physiological pH. This property allows NP10679 to act selectively in ischemic tissue while minimizing the nonselective blockade of NMDA receptors in healthy brain, thereby reducing on-target adverse effects. We report the results of a first-in-human pharmacokinetic and safety phase 1 clinical trial in healthy volunteers receiving single or multiple doses of NP10679 (NCT04007263). We found that NP10679 was well-tolerated and with a half-life of 20 hours, which is amenable to once per day dosing. The only notable side effect in this clinical trial was modest somnolence at higher doses, atypical in that the subject could easily be aroused. The overall results suggest that NP10679 is a candidate for further development for use in acute brain injury, such as ischemic stroke or aneurysmal subarachnoid hemorrhage, as well as for use in neuropsychiatric indications.
Collapse
Affiliation(s)
| | - Stephen F. Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, Atlanta GA 30322
| | - Ray Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, Atlanta GA 30322
| | | | - Daniel T. Laskowitz
- Department of Neurology, Duke University, Bryan Research Building, Durham, NC 27701
| |
Collapse
|
2
|
Yang Y, Gao H, Liu W, Jiang X, Shen Z, Li X, Ren T, Xu Z, Cheng G, Zhao Q. DCMQA, a caffeoylquinic acid derivative alleviates NMDA-induced neurotoxicity via modulating GluN2A and GluN2B-containing NMDA receptors in vitro. Toxicol In Vitro 2020; 67:104888. [PMID: 32416136 DOI: 10.1016/j.tiv.2020.104888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022]
Abstract
Compound DCMQA (4, 5-O-dicaffeoyl-1-O-[4-malic acid methyl ester]-quinic acid) is a natural caffeoylquinic acid derivative isolated from Arctium lappa L. roots. Caffeoylquinic acid derivatives have been reported to possess neuroprotective effects through inhibiting oxidative stress and apoptosis in vitro. However, whether DCMQA exerts protective effects on N-methyl-D-aspartate (NMDA)-induced neurotoxicity and the underlying mechanism has not been elucidated. In this study, the results indicated that pretreatment of DCMQA prevented the loss of cell viability and attenuated the LDH leakage in SH-SY5Y cells exposed to NMDA. Hoechst 33342 staining and Annexin V-PI double staining illustrated that DCMQA suppressed NMDA-induced morphological damage and neuronal apoptosis. Moreover, DCMQA inhibited NMDA-mediated Ca2+ influx, excessive intracellular ROS generation and loss of mitochondrial membrane potential (MMP). Western blot analysis showed that DCMQA attenuated the Bax/Bcl-2 ratio, release of cytochrome c as well as expression of caspase-9 and caspase-3. Besides, DCMQA down-regulated GluN2B-containing NMDA receptors (NMDARs) and up-regulated GluN2A-containing NMDARs, promoted the disruption of nNOS and PSD95 as well as activation of CaMK II-α. Furthermore, computational docking study indicated that DCMQA possessed a good affinity for NMDARs. These results indicated that DCMQA protects SH-SY5Y cells against NMDA-induced neuronal damage. In addition, the underlying mechanisms of DCMQA-mediated neuroprotection are associated with modulating NMDARs and disruption of nNOS-PSD95 as well as the activation of CaMK II-α.
Collapse
Affiliation(s)
- Yue Yang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Huan Gao
- Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Wenwu Liu
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Xiaowen Jiang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Zexu Shen
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Xiang Li
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Tianshu Ren
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Zihua Xu
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Gang Cheng
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China
| | - Qingchun Zhao
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning 110016, People's Republic of China.
| |
Collapse
|
3
|
PKCγ and PKCε are Differentially Activated and Modulate Neurotoxic Signaling Pathways During Oxygen Glucose Deprivation in Rat Cortical Slices. Neurochem Res 2019; 44:2577-2589. [PMID: 31541352 DOI: 10.1007/s11064-019-02876-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 10/26/2022]
Abstract
Cerebral ischemia is known to trigger a series of intracellular events such as changes in metabolism, membrane function and intracellular transduction, which eventually leads to cell death. Many of these processes are mediated by intracellular signaling cascades that involve protein kinase activation. Among all the kinases activated, the serine/threonine kinase family, protein kinase C (PKC), particularly, has been implicated in mediating cellular response to cerebral ischemic and reperfusion injury. In this study, using oxygen-glucose deprivation (OGD) in acute cortical slices as an in vitro model of cerebral ischemia, I show that PKC family of isozymes, specifically PKCγ and PKCε are differentially activated during OGD. Detecting the expression and activation levels of these isozymes in response to different durations of OGD insult revealed an early activation of PKCε and delayed activation of PKCγ, signifying their roles in response to different durations and stages of ischemic stress. Specific inhibition of PKCγ and PKCε significantly attenuated OGD induced cytotoxicity, rise in intracellular calcium, membrane depolarization and reactive oxygen species formation, thereby enhancing neuronal viability. This study clearly suggests that PKC family of isozymes; specifically PKCγ and PKCε are involved in OGD induced intracellular responses which lead to neuronal death. Thus isozyme specific modulation of PKC activity may serve as a promising therapeutic route for the treatment of acute cerebral ischemic injury.
Collapse
|
4
|
Kumagai A, Sasaki T, Matsuoka K, Abe M, Tabata T, Itoh Y, Fuchino H, Wugangerile S, Suga M, Yamaguchi T, Kawahara H, Nagaoka Y, Kawabata K, Furue MK, Takemori H. Monitoring of glutamate-induced excitotoxicity by mitochondrial oxygen consumption. Synapse 2018; 73:e22067. [PMID: 30120794 DOI: 10.1002/syn.22067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 12/14/2022]
Abstract
Dysfunction of mitochondrial activity is often associated with the onset and progress of neurodegenerative diseases. Membrane depolarization induced by Na+ influx increases intracellular Ca2+ levels in neurons, which upregulates mitochondrial activity. However, overlimit of Na+ influx and its prolonged retention ultimately cause excitotoxicity leading to neuronal cell death. To return the membrane potential to the normal level, Na+ /K+ -ATPase exchanges intracellular Na+ with extracellular K+ by consuming a large amount of ATP. This is a reason why mitochondria are important for maintaining neurons. In addition, astrocytes are thought to be important for supporting neighboring neurons by acting as energy providers and eliminators of excessive neurotransmitters. In this study, we examined the meaning of changes in the mitochondrial oxygen consumption rate (OCR) in primary mouse neuronal populations. By varying the medium constituents and using channel modulators, we found that pyruvate rather than lactate supported OCR levels and conferred on neurons resistance to glutamate-mediated excitotoxicity. Under a pyruvate-restricted condition, our OCR monitoring could detect excitotoxicity induced by glutamate at only 10 μM. The OCR monitoring also revealed the contribution of the N-methyl-D-aspartate receptor and Na+ /K+ -ATPase to the toxicity, which allowed evaluating spontaneous excitation. In addition, the OCR monitoring showed that astrocytes preferentially used glutamate, not glutamine, for a substrate of the tricarboxylic acid cycle. This mechanism may be coupled with astrocyte-dependent protection of neurons from glutamate-mediated excitotoxicity. These results suggest that OCR monitoring would provide a new powerful tool to analyze the mechanisms underlying neurotoxicity and protection against it.
Collapse
Affiliation(s)
- Ayako Kumagai
- Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Life Science and Biotechnology, Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenta Matsuoka
- Laboratory for Biological Information Processing, Graduate School of, Science and Engineering, University of Toyama, Toyama, Japan
| | - Masayoshi Abe
- Laboratory for Biological Information Processing, Graduate School of, Science and Engineering, University of Toyama, Toyama, Japan
| | - Toshihide Tabata
- Laboratory for Biological Information Processing, Graduate School of, Science and Engineering, University of Toyama, Toyama, Japan
| | - Yumi Itoh
- Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hiroyuki Fuchino
- Research Center for Medicinal Plant Resources, Tukuba Division, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Japan
| | - Sartagul Wugangerile
- Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Mika Suga
- Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Tomoko Yamaguchi
- Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hidehisa Kawahara
- Life Science and Biotechnology, Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | - Yasuo Nagaoka
- Life Science and Biotechnology, Chemistry, Materials and Bioengineering, Kansai University, Osaka, Japan
| | - Kenji Kawabata
- Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Miho Kusuda Furue
- Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hiroshi Takemori
- Cell Signaling and Metabolic Disease, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Faculty of Engineering, Depaetment of Chemistry and Biomolecular Science, Gifu University, Gifu, Japan
| |
Collapse
|
5
|
Johnson NW, Özkan M, Burgess AP, Prokic EJ, Wafford KA, O'Neill MJ, Greenhill SD, Stanford IM, Woodhall GL. Phase-amplitude coupled persistent theta and gamma oscillations in rat primary motor cortex in vitro. Neuropharmacology 2017; 119:141-156. [PMID: 28400257 DOI: 10.1016/j.neuropharm.2017.04.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/26/2023]
Abstract
In vivo, theta (4-7 Hz) and gamma (30-80 Hz) neuronal network oscillations are known to coexist and display phase-amplitude coupling (PAC). However, in vitro, these oscillations have for many years been studied in isolation. Using an improved brain slice preparation technique we have, using co-application of carbachol (10 μM) and kainic acid (150 nM), elicited simultaneous theta (6.6 ± 0.1 Hz) and gamma (36.6 ± 0.4 Hz) oscillations in rodent primary motor cortex (M1). Each oscillation showed greatest power in layer V. Using a variety of time series analyses we detected significant cross-frequency coupling in 74% of slice preparations. Differences were observed in the pharmacological profile of each oscillation. Thus, gamma oscillations were reduced by the GABAA receptor antagonists, gabazine (250 nM and 2 μM), and picrotoxin (50 μM) and augmented by AMPA receptor antagonism with SYM2206 (20 μM). In contrast, theta oscillatory power was increased by gabazine, picrotoxin and SYM2206. GABAB receptor blockade with CGP55845 (5 μM) increased both theta and gamma power, and similar effects were seen with diazepam, zolpidem, MK801 and a series of metabotropic glutamate receptor antagonists. Oscillatory activity at both frequencies was reduced by the gap junction blocker carbenoxolone (200 μM) and by atropine (5 μM). These data show theta and gamma oscillations in layer V of rat M1 in vitro are cross-frequency coupled, and are mechanistically distinct. The development of an in vitro model of phase-amplitude coupled oscillations will facilitate further mechanistic investigation of the generation and modulation of coupled activity in mammalian cortex.
Collapse
Affiliation(s)
- Nicholas W Johnson
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom
| | - Mazhar Özkan
- Department of Anatomy, School of Medicine, Marmara University, Istanbul, Turkey
| | - Adrian P Burgess
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom
| | - Emma J Prokic
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom
| | - Keith A Wafford
- Neuroscience Division, Eli Lilly & Co. Ltd., Windlesham, GU20 6PH, United Kingdom
| | - Michael J O'Neill
- Neuroscience Division, Eli Lilly & Co. Ltd., Windlesham, GU20 6PH, United Kingdom
| | - Stuart D Greenhill
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom
| | - Ian M Stanford
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom
| | - Gavin L Woodhall
- Aston Brain Centre, Aston University, School of Life and Health Sciences, Birmingham, B4 7ET, United Kingdom.
| |
Collapse
|
6
|
Hansen KB, Yi F, Perszyk RE, Menniti FS, Traynelis SF. NMDA Receptors in the Central Nervous System. Methods Mol Biol 2017; 1677:1-80. [PMID: 28986865 DOI: 10.1007/978-1-4939-7321-7_1] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NMDA-type glutamate receptors are ligand-gated ion channels that mediate a major component of excitatory neurotransmission in the central nervous system (CNS). They are widely distributed at all stages of development and are critically involved in normal brain functions, including neuronal development and synaptic plasticity. NMDA receptors are also implicated in the pathophysiology of numerous neurological and psychiatric disorders, such as ischemic stroke, traumatic brain injury, Alzheimer's disease, epilepsy, mood disorders, and schizophrenia. For these reasons, NMDA receptors have been intensively studied in the past several decades to elucidate their physiological roles and to advance them as therapeutic targets. Seven NMDA receptor subunits exist that assemble into a diverse array of tetrameric receptor complexes, which are differently regulated, have distinct regional and developmental expression, and possess a wide range of functional and pharmacological properties. The diversity in subunit composition creates NMDA receptor subtypes with distinct physiological roles across neuronal cell types and brain regions, and enables precise tuning of synaptic transmission. Here, we will review the relationship between NMDA receptor structure and function, the diversity and significance of NMDA receptor subtypes in the CNS, as well as principles and rules by which NMDA receptors operate in the CNS under normal and pathological conditions.
Collapse
Affiliation(s)
- Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA. .,Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, USA.
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Riley E Perszyk
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., George & Anne Ryan Institute for Neuroscience, Kingston, RI, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
7
|
Raised Intracellular Calcium Contributes to Ischemia-Induced Depression of Evoked Synaptic Transmission. PLoS One 2016; 11:e0148110. [PMID: 26934214 PMCID: PMC4775070 DOI: 10.1371/journal.pone.0148110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/13/2016] [Indexed: 12/26/2022] Open
Abstract
Oxygen-glucose deprivation (OGD) leads to depression of evoked synaptic transmission, for which the mechanisms remain unclear. We hypothesized that increased presynaptic [Ca2+]i during transient OGD contributes to the depression of evoked field excitatory postsynaptic potentials (fEPSPs). Additionally, we hypothesized that increased buffering of intracellular calcium would shorten electrophysiological recovery after transient ischemia. Mouse hippocampal slices were exposed to 2 to 8 min of OGD. fEPSPs evoked by Schaffer collateral stimulation were recorded in the stratum radiatum, and whole cell current or voltage clamp recordings were performed in CA1 neurons. Transient ischemia led to increased presynaptic [Ca2+]i, (shown by calcium imaging), increased spontaneous miniature EPSP/Cs, and depressed evoked fEPSPs, partially mediated by adenosine. Buffering of intracellular Ca2+ during OGD by membrane-permeant chelators (BAPTA-AM or EGTA-AM) partially prevented fEPSP depression and promoted faster electrophysiological recovery when the OGD challenge was stopped. The blocker of BK channels, charybdotoxin (ChTX), also prevented fEPSP depression, but did not accelerate post-ischemic recovery. These results suggest that OGD leads to elevated presynaptic [Ca2+]i, which reduces evoked transmitter release; this effect can be reversed by increased intracellular Ca2+ buffering which also speeds recovery.
Collapse
|
8
|
Yuan H, Myers SJ, Wells G, Nicholson KL, Swanger SA, Lyuboslavsky P, Tahirovic YA, Menaldino DS, Ganesh T, Wilson LJ, Liotta DC, Snyder JP, Traynelis SF. Context-dependent GluN2B-selective inhibitors of NMDA receptor function are neuroprotective with minimal side effects. Neuron 2015; 85:1305-1318. [PMID: 25728572 DOI: 10.1016/j.neuron.2015.02.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/12/2015] [Accepted: 01/29/2015] [Indexed: 01/09/2023]
Abstract
Stroke remains a significant problem despite decades of work on neuroprotective strategies. NMDA receptor (NMDAR) antagonists are neuroprotective in preclinical models, but have been clinically unsuccessful, in part due to side effects. Here we describe a prototypical GluN2B-selective antagonist with an IC50 value that is 10-fold more potent at acidic pH 6.9 associated with ischemic tissue compared to pH 7.6, a value close to the pH in healthy brain tissue. This should maximize neuroprotection in ischemic tissue while minimizing on-target side effects associated with NMDAR blockade in noninjured brain regions. We have determined the mechanism underlying pH-dependent inhibition and demonstrate the utility of this approach in vivo. We also identify dicarboxylate dimers as a novel proton sensor in proteins. These results provide insight into the molecular basis of pH-dependent neuroprotective NMDAR block, which could be beneficial in a wide range of neurological insults associated with tissue acidification.
Collapse
Affiliation(s)
- Hongjie Yuan
- Department of Pharmacology, Emory University, Atlanta, GA 30322 USA
| | - Scott J Myers
- Department of Pharmacology, Emory University, Atlanta, GA 30322 USA
| | - Gordon Wells
- Department of Chemistry, Emory University, Atlanta, GA 30322 USA
| | - Katherine L Nicholson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298 USA.,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Sharon A Swanger
- Department of Pharmacology, Emory University, Atlanta, GA 30322 USA
| | | | | | | | - Thota Ganesh
- Department of Pharmacology, Emory University, Atlanta, GA 30322 USA
| | | | - Dennis C Liotta
- Department of Chemistry, Emory University, Atlanta, GA 30322 USA
| | - James P Snyder
- Department of Chemistry, Emory University, Atlanta, GA 30322 USA
| | | |
Collapse
|
9
|
Kannurpatti SS, Sanganahalli BG, Mishra S, Joshi PG, Joshi NB. Glutamate-induced differential mitochondrial response in young and adult rats. Neurochem Int 2004; 44:361-9. [PMID: 14643754 DOI: 10.1016/s0197-0186(03)00164-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Excitatory amino acid glutamate is involved in neurotransmission in the nervous system but it becomes a potent neurotoxin under variety of conditions. However, the molecular mechanism of excitotoxicity is not known completely. We have studied the influence of glutamate on intracellular calcium and mitochondrial functions in cortical slices from young and adult rats. The slices from both the age groups exhibited comparable intracellular calcium changes upon glutamate stimulation. Glutamate treatment caused a decrease in adenosine 5'-diphosphate/adenosine 5'-triphosphate (ADP/ATP) and an increase in nicotinamide adenine dinucleotide/nicotinamide adenine dinucleotide reduced form (NAD/NADH) ratio in both the age groups but the magnitude and the nature of temporal change was different. Glutamate-induced decrease in ATP/ADP and increase in NAD/NADH ratio was significantly higher in slices from the adult as compared to the young rats. The slices from young rats elicited slightly higher mitochondrial depolarization than adult rats. However, the formation of reactive oxygen species (ROS) and lactate dehydrogenase (LDH) release were significantly higher in adult rats as compared to young rats. Glutamate-induced mitochondrial depolarization, ROS formation and LDH release were highly dependent on the presence of Ca(2+) in the extracellular medium. The treatment of slices with mitochondrial inhibitors rotenone and oligomycin inhibited ROS formation and LDH release substantially. Our results suggest that the glutamate-induced increase in intracellular calcium is not the only factor responsible for neuronal cell death but the mitochondrial functions could be crucial in excitotoxicity.
Collapse
Affiliation(s)
- S S Kannurpatti
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences, Bangalore 560 029, India
| | | | | | | | | |
Collapse
|
10
|
Lehmann A, Jacobson I. Ion Dependence and Receptor Mediation of Glutamate Toxicity in the Immature Rat Hippocampal Slice. Eur J Neurosci 2002; 2:620-628. [PMID: 12106296 DOI: 10.1111/j.1460-9568.1990.tb00451.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Glutamate (glu) is a major excitatory transmitter and a toxin in the brain. In the present study, the immature rat hippocampal slice was used to determine the morphology, topography, ionic mediation and receptor specificity of glu toxicity. Slices were exposed to glu for 30 min, and the damage was evaluated after 3 h of recovery in regular medium. The effects on glu toxicity of changes of [Ca2+], [Cl-] and [Na+] were determined. The receptor preference of glu was assessed by using the N-methyl-D-aspartate (NMDA) antagonist MK-801 and the kainate (KA)/quisqualate (QA) antagonist DNQX, alone or in combination. Further, to see whether glu produces cytotoxicity via osmolysis, the effects of hyperosmolal sucrose on glu toxicity were studied. Glu toxicity was similar to the previously described NMDA toxicity with regard to cytopathology, but differed in some aspects from that caused by KA and QA. The severity of the lesion was determined by the proximity of neurons to the incubation fluid, probably as a consequence of cellular accumulation of the amino acid. Omission of Ca2+ abolished glu toxicity in all neurons except the granule cells of the outer blade. This population was completely protected when Ca2+ was omitted and [Cl-] was reduced. Elevation of [Ca2+] markedly aggravated the lesion caused by glu. Substitution of isethionate for Cl- worsened the glu-induced damage, whilst the amino acid produced qualitatively different neuropathology when choline substituted for Na+. Apparently glu did not damage hippocampal nerve cells through an osmolytic mechanism as medium supplemented with 100 mM sucrose increased the toxicity of glu. Since the lesion produced by glu was more widespread in the presence of high [Ca2+], the effects of receptor antagonists were studied under this condition. MK-801 inhibited glu toxicity whereas DNQX had no effect. Combination of MK-801 and DNQX did not offer better protection than did MK-801 alone. The results suggest that Ca2+ is the main (but not single) determinant of glu toxicity in the immature hippocampal slice. The ionic requirements of glu neurotoxicity are identical to those of NMDA, but differ from those of KA and QA. The notion that glu is a selective NMDA agonist in the present model was confirmed by the protection of MK-801, and by the lack of an effect of DNQX. This is the first report demonstrating that the toxicity of glu is mediated by NMDA receptors in brain tissue which has developed normally. The findings indicate that specific blockade of NMDA receptors may be the most rational strategy in the prevention of glu-related neuronal death occurring in certain neurological anomalies.
Collapse
Affiliation(s)
- Anders Lehmann
- Institute of Neurobiology, University of Göteborg, P.O. Box 33031, S-400 33 Göteborg, Sweden
| | | |
Collapse
|
11
|
Abstract
The mechanisms underlying the neurodegenerative effects of the glutamate receptor agonist, AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate), were studied using brain slice preparations of young rat (8 - 9 days old) cerebellum and hippocampus. Rapid AMPA toxicity (exerted on some cerebellar interneurons) was inhibited by including the appropriate receptor blocker, CNQX (6-cyano-7-nitroquinoxaline-2,3-dione, 10 microM), in the exposing solution. The degeneration of other neurons, including Purkinje cells and hippocampal pyramidal neurons, persisted. It could, however, be largely prevented if CNQX was included for 1.5 h during the post-incubation period, suggesting that an enduring 'rebound' AMPA receptor activation was responsible for this delayed type of degeneration, not the exposure itself. In cerebellar slices, independent evidence for the occurrence, postexposure, of persisting AMPA receptor stimulation was obtained electrophysiologically. Omission of Ca2+ during the exposure period (and for 10 min beforehand) markedly reduced rapid AMPA toxicity but was ineffective in protecting most of the Purkinje cells. However, if the slices were previously starved of Ca2+ for 1 h, then most of these neurons survived, even if the ion was reinstated during the recovery period. Slow AMPA toxicity, which takes place during long (2 h) exposures, could be inhibited either by CNQX or by omission of Ca2+ (30 min preincubation). The results indicate that the rapid oedematous necrosis induced by AMPA, like that caused by N-methyl-d-aspartate and kainate, is likely to involve excessive influx of Ca2+. In contrast, the induction of the delayed mechanisms, as well as its 'expression' during the postincubation period, probably depends on intracellular Ca2+, rather than Ca2+ influx.
Collapse
Affiliation(s)
- Giti Garthwaite
- Department of Physiology, University of Liverpool, Brownlow Hill, PO Box 147, Liverpool L69 3BX, UK
| | | |
Collapse
|
12
|
Hafidi A, Lanjun G, Sanes DH. Age-dependent failure of axon regeneration in organotypic culture of gerbil auditory midbrain. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(19991105)41:2<267::aid-neu9>3.0.co;2-d] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
13
|
Kimura M, Katayama K, Nishizawa Y. Role of glutamate receptors and voltage-dependent calcium channels in glutamate toxicity in energy-compromised cortical neurons. JAPANESE JOURNAL OF PHARMACOLOGY 1999; 80:351-8. [PMID: 10496336 DOI: 10.1254/jjp.80.351] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We have examined the effect of glutamate receptor antagonists and voltage-dependent calcium channel blockers on the neuronal injury induced by the combination of a low concentration of N-methyl-D-aspartate (NMDA) or kainate and energy compromise resulting from the use of glucose-free incubation buffer. Toxicity induced by NMDA or kainate was enhanced in the glucose-free buffer. NMDA-or non-NMDA-receptor antagonists added to the glucose-free buffer at the same time inhibited the neuronal cell death induced by each agonist. An NMDA-receptor antagonist, MK-801, but not non-NMDA-receptor antagonists, inhibited the toxicity when added to the culture medium after exposure of the cells to the agonists. P/Q-type calcium channel blockers, omega-agatoxin IVA and omega-agatoxin TK, and an N-type calcium channel blocker, omega-conotoxin GVIA, significantly attenuated the neuronal injury, although an L-type calcium channel blocker, nifedipine, showed little neuroprotective effect. A combination of calcium channel blockers of the three subtypes showed the most prominent neuroprotective effect. These observations suggest that the overactivation of NMDA and non-NMDA receptors and consequent activation of the voltage-dependent calcium channels lead to neuronal cell death in energy-compromised cortical neurons.
Collapse
Affiliation(s)
- M Kimura
- Eisai Tsukuba Research Laboratories, Ibaraki, Japan
| | | | | |
Collapse
|
14
|
Abstract
Ascorbate and glutathione (GSH) are normally concentrated in brain cells at millimolar levels. However, both of these low-molecular-weight antioxidants are washed out of mammalian brain tissue during slice preparation and subsequent incubation. Ascorbate, which is not synthesized in the brain, can be added back to slices by active uptake from the incubation medium. Levels of GSH, on the other hand, are regulated by synthesis rather than uptake, and cannot be readily maintained in slices. Importantly, maintenance of brain slice ascorbate content at at least 50% of that in vivo, prevents the increase in slice water content that normally occurs during incubation. Slices with maintained ascorbate levels also have better histological characteristics than ascorbate-depleted tissue. The medium concentration of ascorbate sufficient to maintain content and inhibit edema formation is 400 microM, which is the normal concentration in brain extracellular fluid. This paper describes methods to maintain ascorbate levels in brain slices, including procedures to minimize oxidation in oxygenated incubation media. Also described is an HPLC analysis for ascorbate and GSH that is based on direct injection rather than extraction of samples.
Collapse
Affiliation(s)
- M E Rice
- Departments of Neurosurgery and Physiology and Neuroscience, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA.
| |
Collapse
|
15
|
Safran N, Haring R, Shainberg A, Zisling R, Futerman AH, Shahar A. Nerve cell death induced by Ca2+ ionophores in dissociated hippocampal cultures. Protective action of the NMDA antagonist MK-801. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1997; 429:207-19. [PMID: 9413576 DOI: 10.1007/978-1-4757-9551-6_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- N Safran
- Koret School of Vet. Med., Hebrew University of Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
16
|
Liu S, Ruenes GL, Yezierski RP. NMDA and non-NMDA receptor antagonists protect against excitotoxic injury in the rat spinal cord. Brain Res 1997; 756:160-7. [PMID: 9187327 DOI: 10.1016/s0006-8993(97)00137-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The neuroprotective properties of the N-methyl-D-aspartate (NMDA) antagonist dizocilpine (MK-801) and the non-NMDA antagonists 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f]quinoxaline (NBQX) and alpha-methyl-4-carboxyphenylglycine (MCPG) were evaluated against neuronal injury produced by the intraspinal injection of NMDA and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA). Forty-nine animals were divided into eight groups in order to evaluate the effects of different drug combinations: (a) NMDA; (b) NMDA + MCPG; (c) NMDA + NBQX; (d) NMDA + MK-801; (e) AMPA; (f) AMPA + MCPG; (g) AMPA + MK-801; and (h) AMPA + NBQX. Drugs were microinjected into spinal segments T12-L3 through a micropipette attached to a Hamilton microliter syringe. Spinal cords were evaluated after a survival period of 48 h at which time NMDA and AMPA were found to produce morphological changes over the concentration ranges of 125-500 mM and 75-500 microM, respectively. Neuronal loss following injections of NMDA + MK-801 or AMPA + NBQX was significantly less than that following injections of NMDA or AMPA alone. By contrast, neuronal loss following co-injections of NMDA or AMPA with inappropriate antagonists, i.e., NMDA + NBQX/MCPG or AMPA + MCPG/MK-801, was not significantly different from that produced by NMDA or AMPA. The results suggest that elevations in spinal levels of glutamate followed by prolonged activation of NMDA and AMPA receptor subtypes initiate an excitotoxic cascade resulting in neuronal injury. Blockade of NMDA and AMPA effects by MK-801 and NBQX respectively confirms the well documented neuroprotective effects of these drugs and lends support to the potential importance of NMDA and especially AMPA receptor antagonists as therapeutic agents in the treatment of acute spinal cord injury.
Collapse
Affiliation(s)
- S Liu
- The Miami Project to Cure Paralysis, University of Miami, FL 33136, USA
| | | | | |
Collapse
|
17
|
Tymianski M, Tator CH. Normal and abnormal calcium homeostasis in neurons: a basis for the pathophysiology of traumatic and ischemic central nervous system injury. Neurosurgery 1996; 38:1176-95. [PMID: 8727150 DOI: 10.1097/00006123-199606000-00028] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Clinical recovery after central nervous system (CNS) trauma or ischemia may be limited by a neural injury process that is triggered and perpetuated at the cellular level, rather than by a lesion amenable to surgical repair. It is widely thought that one such process, a fundamental pathological mechanism initiated by CNS injury, is a disruption of cellular Ca2+ homeostasis. Because of the critical role of Ca2+ ions in regulating innumerable cellular functions, this major homeostatic disturbance is thought to trigger neuronal and axonal degeneration and produce clinical disability. We review those aspects of normal and pathological Ca2+ homeostasis in neurons that relate to neurodegeneration and to the application of neuroprotective strategies for the treatment of CNS injury. In particular, we examine the contribution of Ca(2+)-permeable ionic channels, Ca2+ pumps, intracellular Ca2+ stores, intracellular Ca2+ buffering systems, and the roles of secondary, Ca(2+)-dependent processes in neurodegeneration. A number of hypotheses linking Ca2+ ions and Ca2+ permeable channels to neurotoxicity are discussed with an emphasis on strategies for lessening Ca(2+)-related damage. A number of these strategies may have a future role in the treatment of traumatic and ischemic CNS injury.
Collapse
Affiliation(s)
- M Tymianski
- Division of Neurosurgery, Toronto Hospital, Ontario, Canada
| | | |
Collapse
|
18
|
Tymianski M, Tator CH. Normal and Abnormal Calcium Homeostasis in Neurons: A Basis for the Pathophysiology of Traumatic and Ischemic Central Nervous System Injury. Neurosurgery 1996. [DOI: 10.1227/00006123-199606000-00028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
19
|
Boireau A, Malgouris C, Burgevin MC, Pény C, Durand G, Bordier F, Meunier M, Miquet JM, Daniel M, Chevet T, Jimonet P, Mignani S, Blanchard JC, Doble A. Neuroprotective effects of RPR 104632, a novel antagonist at the glycine site of the NMDA receptor, in vitro. Eur J Pharmacol 1996; 300:237-46. [PMID: 8739214 DOI: 10.1016/0014-2999(95)00780-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The NMDA antagonist and neuroprotective effects of RPR 104632 (2H-1,2,4-benzothiadiazine-1-dioxide-3-carboxylic acid), a new benzothiadiazine derivative, with affinity for the glycine site of the NMDA receptor-channel complex are described. RPR 104632 antagonized the binding of [3H]5,7-dichlorokynurenic acid to the rat cerebral cortex, with a Ki of 4.9 nM. This effect was stereospecific, since the (-)-isomer was 500-fold more potent than the (+)-isomer. The potent affinity of RPR 104632 for the glycine site was confirmed by the observation that RPR 104632 inhibited [3H]N-[1-(2-thienyl)cyclohexyl]-3,4-piperidine ([3H]TCP) binding in the presence of N-methyl-D-aspartate (NMDA) (IC50 = 55 nM), whereas it had no effect on the competitive NMDA site or on the dissociative anaesthetic site. RPR 104632 inhibited the NMDA-evoked increase in guanosine 3',5'-cyclic monophosphate (cGMP) levels of neonatal rat cerebellar slices (IC50 = 890 nM) in a non-competitive manner and markedly reduced NMDA-induced neurotoxicity in rat hippocampal slices and in cortical primary cell cultures. These results suggest that RPR 104632 is a high-affinity specific antagonist of the glycine site coupled to the NMDA receptor channel with potent neuroprotective properties in vitro.
Collapse
Affiliation(s)
- A Boireau
- Rhône Poulenc Rorer, Centre de Recherche de Vitry-Alfortville, Vitry-sur-Seine, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tymianski M, Spigelman I, Zhang L, Carlen PL, Tator CH, Charlton MP, Wallace MC. Mechanism of action and persistence of neuroprotection by cell-permeant Ca2+ chelators. J Cereb Blood Flow Metab 1994; 14:911-23. [PMID: 7929656 DOI: 10.1038/jcbfm.1994.122] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cell-permeant Ca2+ chelators such as 1,2-bis-(2-aminophenoxy)ethane- N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM) have been reported to protect neurons in experimental focal cerebral ischemia. However, their in vivo actions are uncertain, and their protective efficacy is proven only in brief cerebral ischemia paradigms. Here we examine their mechanism of action in vitro and duration of efficacy in vivo. Electrophysiological studies were made in CA1 neurons in rat hippocampal slices. When superfused with BAPTA-AM (30-50 microM), CA1 somatic field potential recordings showed attenuation of the population spike amplitude, and intracellular recordings showed reduced excitatory postsynaptic potentials, indicating inhibition of excitatory synaptic transmission. Also, Ca(2+)-dependent accommodation and post-spike-train hyperpolarizations were reduced, indicating Ca2+ chelation hear the internal cell membrane surface. To determine whether Ca2+ chelators reduce the size of cerebral infarction rather than simply delaying its evolution, we studied the effects of BAPTA-AM treatment on infarction size 24 h after permanent middle cerebral artery occlusion. Fischer rats (n = 8 per group) were pretreated with saline, BAPTA-AM (20 mg/kg), or MK-801 (0.5 mg/kg). Infarction volumes in animals treated with BAPTA-AM were reduced by 50.5% compared with controls (p = 0.018), whereas animals treated with MK-801 experienced a statistically insignificant infarct volume reduction (26%; p = 0.27). These data show a persistence of neuroprotection by the Ca2+ chelator at 24 h and indicate that it may act by attenuating synaptic transmission and subplasma membrane Ca2+ excess.
Collapse
Affiliation(s)
- M Tymianski
- Playfair Neuroscience Unit, Toronto Hospital Research Institute, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
21
|
Malgouris C, Daniel M, Doble A. Neuroprotective effects of riluzole on N-methyl-D-aspartate- or veratridine-induced neurotoxicity in rat hippocampal slices. Neurosci Lett 1994; 177:95-9. [PMID: 7824190 DOI: 10.1016/0304-3940(94)90053-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The neuroprotective activity of riluzole has been studied on N-methyl-D-aspartate (NMDA)- or veratridine-induced toxicity in immature rat hippocampal slices. Neurodegeneration was assessed by the measurement of LDH release and histology. Veratridine-induced LDH release can be inhibited by 100 microM riluzole (-90% and by tetrodotoxin (1 microM). Riluzole markedly reduced (-59%) the NMDA-induced LDH release and this protective effect was confirmed by histology. Riluzole inhibited the NMDA-induced LDH release in the presence of tetrodotoxin. Moreover, a pretreatment with pertussis toxin (1 microgram/ml) abolished the effect of riluzole against NMDA-induced neurotoxicity. These results support the view that the neuroprotective properties of riluzole could be exerted via two distinct mechanisms of action.
Collapse
Affiliation(s)
- C Malgouris
- Rhône-Poulenc Rorer, Centre de Recherche de Vitry-Alfortville, Vitry-sur-Seine, France
| | | | | |
Collapse
|
22
|
Morari M, Menegale M, Caló G, Ferraro L, Tomasini C, Bianchi C, Beani L. Excitatory amino acids (EAAs) stimulate phosphatidylinositol turnover in adult rat striatal slices: interaction between NMDA and EAA metabotropic receptors. Neurochem Int 1994; 24:191-200. [PMID: 8161946 DOI: 10.1016/0197-0186(94)90106-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The effect of excitatory amino acids (EAAs) on phosphatidylinositol (PI) turnover in adult rat striatal slices was investigated. Quisqualic acid (QA), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainic acid (KA), ibotenic acid (IBO) and N-methyl-D-aspartic acid (NMDA) maximally increased inositol phosphate (IP) formation at 10 microM while trans-1-amino-cyclopentane-1,3-dicarboxylic acid (ACPD) was maximally effective at 100 microM. The NMDA channel blocker dizolcipine (MK-801) counteracted the effect of NMDA 10 microM and IBO 10 microM while it potentiated that of IBO 100 microM and IBO 1000 microM. Conversely, the non-NMDA receptor antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) prevented the effect of AMPA and KA and reduced that of QA (all at 10 microM). Lowering extracellular Ca2+ concentrations ([Ca2+]0) differentially affected the PI response to EAAs. The ACPD 30 microM effect was unchanged at low [Ca2+]0 (but abolished when EGTA 2 mM was added), while that of ACPD 100 microM was halved in 0.1 mM and almost abolished in a nominally free Ca2+ medium. NMDA 10 microM and AMPA 10 microM were ineffective at low [Ca2+]0 while NMDA 100 microM, ineffective in a 1.2 mM Ca2+ medium, strongly stimulated IP formation in 0.1 mM Ca2+ but not in a nominally free Ca2+ medium. The effect of NMDA on EAA metabotropic receptor agonist stimulated PI turnover was also studied. NMDA 10 microM potentiated the effect of ACPD 30 microM. This positive cooperation persisted at low [Ca2+]0 but not in the presence of EGTA. Conversely, NMDA 100 microM prevented the effect of ACPD 100 microM. This negative interference was reversed when Ca2+ was omitted from the medium. This study shows that in the adult rat striatum both EAA metabotropic and ionotropic receptor activation increases IP formation. A positive and negative interaction between NMDA and metabotropic receptor activation was also found to regulate PI turnover. The role of [Ca2+]0 in subserving the PI response to EAAs was made evident.
Collapse
Affiliation(s)
- M Morari
- Institute of Pharmacology, University of Ferrara, Italy
| | | | | | | | | | | | | |
Collapse
|
23
|
Lehmann A, Hagberg H, Orwar O, Sandberg M. Cysteine sulphinate and cysteate: mediators of cysteine toxicity in the neonatal rat brain? Eur J Neurosci 1993; 5:1398-412. [PMID: 7903894 DOI: 10.1111/j.1460-9568.1993.tb00926.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Excitotoxic amino acids contain two acidic groups, but cysteine represents an exception to this rule. The hypothesis that cysteine toxicity is mediated by the oxidized and diacidic metabolites cysteine sulphinate and/or cysteate was tested in the present study. The issue was approached in three different ways. Firstly, the distribution of brain injury after subcutaneous administration of cysteine (1 mg/g) to 4-day-old rats was compared with that caused by cysteine sulphinate (3 mg/g). Secondly, the effects of excitatory amino acid receptor antagonists on cysteine and cysteine sulphinate toxicity were investigated. Thirdly, the cerebral concentrations of cysteine sulphinate were determined after cysteine administration and compared with those obtained after cysteine sulphinate injection. The cerebral cortex was the region most vulnerable to cysteine toxicity, followed by the hippocampus (especially the medial subicular neurons), amygdala, caudoputamen, cerebellum and septum. Pronounced extravasation of red blood cells was observed in lesioned areas. One day after cysteine administration, the injury was infarction-like and sharply demarcated. Cysteine sulphinate-induced damage resembled cysteine-induced lesions in some respects: the anterior cingulate and retrosplenial cortices, as well as medial subicular cells, were quite vulnerable. However, the differences prevailed. Cysteine sulphinate, but not cysteine, killed neurons of the superficial part of the tectum, the medial habenula, the ventromedial hypothalamus and the arcuate nucleus. Further, while cysteine toxicity was prominent in deep cortical layers, cysteine sulphinate preferentially damaged superficial cortical neurons. Cysteine toxicity was abolished by pretreatment with MK-801, a selective NMDA antagonist, but not by 2,3-dihydroxy-6-nitro-7-sulphamoyl-benzo(F)quinoxaline, a selective AMPA receptor blocker. In contrast, the considerably smaller lesion seen after cysteine sulphinate administration was only partially prevented by MK-801. Large (19-fold) increases in cortical cysteine sulphinate concentration were noted after injection of a toxic dose of cysteine. This corresponds to 90 nmol cysteine sulphinate/g protein. The cysteate concentration was not increased above the detection limit. Injection of a toxic dose of cysteine sulphinate elevated cysteine sulphinate concentration in the frontomedial cortex (a region consistently injured by cysteine sulphinate) almost three orders of magnitude more than that observed after cysteine administration. Taken together, these results strongly suggest that neither cysteine sulphinate nor cysteate alone mediate cysteine toxicity.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- A Lehmann
- Department of Anatomy and Cell Biology, University of Göteborg, Sweden
| | | | | | | |
Collapse
|
24
|
Menéndez N, Solís JM, Herreras O, Galarreta M, Conejero C, Martín del Río R. Taurine release evoked by NMDA receptor activation is largely dependent on calcium mobilization from intracellular stores. Eur J Neurosci 1993; 5:1273-9. [PMID: 8275229 DOI: 10.1111/j.1460-9568.1993.tb00912.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
It is known that the activation of N-methyl-D-aspartate (NMDA) receptors leads to an increase in extracellular taurine concentration in different brain regions. The mechanism that mediates this effect is not totally understood. In this study, rat hippocampal slices were used to determine the dependence of NMDA-induced taurine release on extracellular calcium and/or on calcium mobilization from intracellular stores. NMDA was administered through a microdialysis probe inserted into the slice, at the level of CA1 stratum radiatum, which was also used to collect amino acids from the extracellular space. Field potentials evoked by stimulation of the Schaffer collaterals and recorded in the stratum pyramidale of CA1 were used as a control of NMDA receptor activation. NMDA induced a marked increase in extracellular taurine levels and a decrease in field potential amplitude, and both effects were suppressed in the presence of MK-801, a blocker of the NMDA receptor-linked channel. Dantrolene, an inhibitor of calcium release from intracellular stores, partially inhibited the extracellular taurine increase, while 2-nitro-4-carboxyphenyl-N,N-diphenyl carbamate (NCDC), an inhibitor of phosphatidylinositol-specific phospholipase C activation, had no effect. Removal of extracellular calcium diminished, but did not abolish, the extracellular taurine increase caused by NMDA. The remaining taurine response was totally suppressed by dantrolene, and also by NCDC. These results demonstrate that the release of taurine induced by NMDA receptor activation is triggered by the increase in cytoplasmic calcium concentration. We suggest that, under physiological conditions, calcium influx provides the signal for NMDA-induced taurine release, which is amplified by calcium-dependent calcium mobilization from intracellular stores.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- N Menéndez
- Departamento de Investigación, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
25
|
Turetsky DM, Huettner JE, Gottlieb DI, Goldberg MP, Choi DW. Glutamate receptor-mediated currents and toxicity in embryonal carcinoma cells. JOURNAL OF NEUROBIOLOGY 1993; 24:1157-69. [PMID: 7691990 DOI: 10.1002/neu.480240904] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
While primary neuronal cell cultures have been used to investigate excitotoxicity, development of cell lines exhibiting glutamate receptor-mediated death is desirable. P19 mouse embryonal carcinoma cells, exposed to retinoic acid and plated onto a layer of cultured mouse cortical glial cells, differentiated into neuron-like elements immunoreactive for neurofilaments and neuron-specific enolase. Whole-cell recordings revealed inward currents in response to extracellular application of either NMDA or kainate. The NMDA-induced currents exhibited a voltage-dependent blockade by magnesium, required glycine for maximal activation, and were blocked by the NMDA antagonist dizocilpine. Kainate-induced currents were blocked by the AMPA/kainate receptor antagonist CNQX. Exposure to 500 microM NMDA for 24 h destroyed most P19 cells (EC50 approximately 70 microM); death was prevented by dizocilpine or D-APV. Exposure to 500 microM kainate also resulted in widespread death reduced by CNQX. Thus differentiated P19 cells exhibited both excitatory amino acid responses and vulnerability to excitotoxicity, characteristic of CNS neurons. These cells may provide a genetically open system useful for studying glutamate receptor-mediated phenomena at a molecular level.
Collapse
Affiliation(s)
- D M Turetsky
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | | | | | |
Collapse
|
26
|
Tymianski M, Charlton MP, Carlen PL, Tator CH. Secondary Ca2+ overload indicates early neuronal injury which precedes staining with viability indicators. Brain Res 1993; 607:319-23. [PMID: 7683241 DOI: 10.1016/0006-8993(93)91523-u] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Spinal neurons, lethally challenged with excitatory amino acids (EAAs) or with high-K+, underwent a biphasic rise in free intracellular calcium concentration ([Ca2+]i). In contrast to the initial rise in [Ca2+]i which recovered, the secondary, irreversible [Ca2+]i increase was unaffected by antagonists of EAA receptors or Ca2+ channels. Also, it correlated highly with cell death, but preceded vital staining with trypan blue and ethidium homodimer, reflecting damaged cellular Ca2+ regulation rather than plasma membrane leakiness. Our findings suggest that delayed Ca2+ overload is the end-product rather than the cause of Ca(2+)-triggered neurotoxic processes.
Collapse
Affiliation(s)
- M Tymianski
- Playfair Neuroscience Unit, Toronto Hospital, Ont., Canada
| | | | | | | |
Collapse
|
27
|
Frandsen A, Schousboe A. Excitatory amino acid-mediated cytotoxicity and calcium homeostasis in cultured neurons. J Neurochem 1993; 60:1202-11. [PMID: 8455022 DOI: 10.1111/j.1471-4159.1993.tb03278.x] [Citation(s) in RCA: 173] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A large body of evidence suggests that disturbances of Ca2+ homeostasis may be a causative factor in the neurotoxicity induced by excitatory amino acids (EAAs). The route or routes by which an increase in intracellular calcium concentration ([Ca2+]i) is mediated in vivo are presently not clarified. This may partly reflect the complexity of intact nervous tissue in combination with the relative unspecific action of the available "calcium antagonists," e.g., blockers of voltage-sensitive calcium channels. By using primary cultures of cortical neurons as a model system, it has been found that all EAAs stimulate increases in [Ca2+]i but via different mechanisms. By using the drug dantrolene, it has been shown that 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propionate (AMPA) apparently exclusively stimulates Ca2+ influx through agonist-operated calcium channels and voltage-operated calcium channels. Increased [Ca2+]i due to exposure to kainate (KA) is for the major part caused by influx, as in the case of AMPA, but a small part of the increase in [Ca2+]i may be attributed to a release of Ca2+ from intracellular stores. Quisqualate (QA) stimulates Ca2+ release from an intracellular store that is independent of Ca2+ influx; presumably this store is activated by inositol phosphates. The increase in [Ca2+]i due to exposure to glutamate or N-methyl-D-aspartate (NMDA) may be compartmentalized into three components, one of which is related to influx and the other two to Ca2+ release from internal stores. Only one of the latter stores is dependent on Ca2+ influx with regard to release of Ca2+, whereas the other is activated by some other second messengers or, alternatively, directly coupled to the receptor. In muscles dantrolene is known to inhibit Ca2+ release from the sarcoplasmic reticulum, and also in neurons dantrolene inhibits an equivalent release from one or more hitherto unidentified internal Ca2+ pool(s). By using this drug it has been possible to show to what extent these Ca2+ stores are involved in the toxicity observed subsequent to exposure to the EAAs. It turned out that dantrolene, even under conditions allowing Ca2+ influx, inhibited toxicity induced by QA, NMDA, and glutamate, whereas that induced by AMPA or KA was unaffected. In combination with the findings that dantrolene inhibited release from the intracellular stores activated by QA, NMDA, and glutamate, it may be concluded that Ca2+ influx per se is not the primary event causing toxicity following exposure to these EAAs in these neurons. However, it may certainly be involved in the cases of toxicity induced by AMPA and KA.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- A Frandsen
- Department of Biological Sciences, Royal Danish School of Pharmacy, Copenhagen
| | | |
Collapse
|
28
|
Frandsen A, Schousboe A, Griffiths R. Cytotoxic actions and effects on intracellular Ca2+ and cGMP concentrations of sulphur-containing excitatory amino acids in cultured cerebral cortical neurons. J Neurosci Res 1993; 34:331-9. [PMID: 8095988 DOI: 10.1002/jnr.490340310] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Effects of the sulphur-containing acidic amino acids (SAAs) cysteic acid (CA), homocysteic acid (HCA), cysteine sulphinic acid (CSA), homocysteine sulphinic acid (HCSA), and S-sulphocysteine (SC) on intracellular concentrations of Ca2+ ([Ca2+]i) and cGMP ([cGMP]i) as well as their cytotoxic actions were investigated in cultured cerebral cortical neurons. The glutamate receptor subtype selective antagonists APV (D-(-)-2-amino-5-phosphonopentanoate) acting on N-methyl-D-aspartate (NMDA) receptors and DNQX (6,7-dinitroquinoxaline-2,3-dione) acting on non-NMDA receptors were employed to obtain information about the involvement of glutamate receptor subtypes in these actions of the SAAs. It was found that all SAAs exerted a cytotoxic action on the neurons. The ED50 values for CSA, CA, HCSA, and HCA were around 30 to 50 microM and that for SC was about 150 microM. The glutamate transport blocker L-aspartate-beta-hydroxamate increased the efficacy of CSA and CA but had no effect on the cytotoxic actions of the remaining SAAs. In case of CA, HCA, and SC the cytotoxicity could be prevented by APV alone and for HCSA, DNQX could block the toxic action. DNQX reduced the toxicity of HCA somewhat but the presence of APV was required for complete protection. CSA toxicity could only be blocked by the combination of APV and DNQX. All SAAs induced an increase in [cGMP]i and [Ca2+]i and with regard to [Ca2+]i SC was the most potent and CA the least potent SAA. The effect of all SAAs on [cGMP]i could be blocked by APV alone whereas DNQX had no effect except in the case of HCSA where the response was blocked completely and HCA where the response was inhibited by 75%.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- A Frandsen
- PharmaBiotec Research Center, Department of Biological Sciences, Royal Danish School of Pharmacy, Copenhagen
| | | | | |
Collapse
|
29
|
Abstract
Excitotoxicity refers to the ability of glutamate or related excitatory amino acids to mediate the death of central neurons under certain conditions, for example, after intense exposure. Such excitotoxic neuronal death may contribute to the pathogenesis of brain or spinal cord injury associated with several human disease states. Excitotoxicity has substantial cellular specificity and, in most cases, is mediated by glutamate receptors. On average, NMDA receptors activation may be able to trigger lethal injury more rapidly than AMPA or kainate receptor activation, perhaps reflecting a greater ability to induce calcium influx and subsequent cellular calcium overload. It is possible that excitotoxic death may share some mechanisms with other forms of neuronal death.
Collapse
Affiliation(s)
- D W Choi
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
30
|
Bohner AP, Dudek SM, Bear MF. Effects of N-methyl-D-aspartate on quisqualate-stimulated phosphoinositide hydrolysis in slices of kitten striate cortex. Brain Res 1992; 594:146-9. [PMID: 1334763 DOI: 10.1016/0006-8993(92)91040-l] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stimulation of phosphoinositide (PI) hydrolysis by excitatory amino acids (EAAs) was studied in coronal slices of kitten visual cortex. Coincubation with N-methyl-D-aspartate (NMDA) markedly reduced the stimulation by quisqualate, however, this inhibition developed with a latency of > 10 min and occurred even when the NMDA exposure preceded, but did not overlap with, incubation in quisqualate. This time-course of NMDA inhibition of EAA-stimulated PI turnover places new constraints on its possible mechanism of inhibition.
Collapse
Affiliation(s)
- A P Bohner
- Department of Neuroscience, Brown University, Providence 02912
| | | | | |
Collapse
|
31
|
Frandsen A, Schousboe A. Mobilization of dantrolene-sensitive intracellular calcium pools is involved in the cytotoxicity induced by quisqualate and N-methyl-D-aspartate but not by 2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl)propionate and kainate in cultured cerebral cortical neurons. Proc Natl Acad Sci U S A 1992; 89:2590-4. [PMID: 1372982 PMCID: PMC48707 DOI: 10.1073/pnas.89.7.2590] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
By using primary cultures of cerebral cortical neurons, it has been demonstrated that the antihyperthermia drug dantrolene protects against cytotoxicity induced by the excitatory amino acids quisqualate (QA) and N-methyl-D-aspartate (NMDA), whereas no effect was observed on cell damage mediated by kainate (KA) or 2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl)propionate (AMPA). In parallel it was shown that KA and AMPA increased the concentration of intracellular free calcium ([Ca2+]i) mainly by influx, whereas the increase in [Ca2+]i stimulated by NMDA and QA predominantly was caused by release of Ca2+ from intracellular stores, which for NMDA seemed to be mediated at least partly by Ca2+ influx. In accordance with the effects on cytotoxicity, dantrolene blocked the increase in [Ca2+]i elicited by QA and NMDA leaving the increase induced by KA and AMPA unaffected. The finding that 2-amino-3-[3-(carboxymethoxy)-5-methylisoxazol-4-yl]propionate, which regarding toxicity is a selective KA antagonist, only reduced the KA-stimulated increase in [Ca2+]i by 30% may suggest that the elevation of [Ca2+]i is not the only element in KA-induced cytotoxicity. On the other hand, the present study underlines the importance of Ca2+ for cytotoxicity induced by some excitatory amino acids (glutamate, NMDA, and QA) and supports the current proposal that multiple mechanisms are operating, even concerning calcium homeostasis. Because excitatory amino acid-induced cytotoxicity is thought to be involved in neuropathological conditions such as ischemia, it is possible that dantrolene might be of therapeutic interest.
Collapse
Affiliation(s)
- A Frandsen
- Department of Biological Sciences, Royal Danish School of Pharmacy, Copenhagen
| | | |
Collapse
|
32
|
Affiliation(s)
- C D Balaban
- Department of Otolaryngology, University of Pittsburgh, Pennsylvania
| | | |
Collapse
|
33
|
Vornov JJ, Coyle JT. Enhancement of NMDA receptor-mediated neurotoxicity in the hippocampal slice by depolarization and ischemia. Brain Res 1991; 555:99-106. [PMID: 1657299 DOI: 10.1016/0006-8993(91)90865-s] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Evidence from animal stroke models suggests that the proximate cause of neuronal degeneration after ischemia is massive release of glutamate and activation of NMDA receptors. However, in the physiologic presence of oxygen and glucose in the rat hippocampal slice preparation, the neurotoxicity of glutamate, as measured by inhibition of protein synthesis, requires high concentrations and is not prevented by glutamate receptor antagonists. Thus, the NMDA receptor-mediated neurotoxic effects of extracellular glutamate accumulation during ischemia might depend on additional factors, such as neuronal depolarization. In the experiments reported here, slices were exposed to glutamate in a medium intended to mimic the ionic conditions found during ischemia, high potassium (128 mM) and low sodium (26 mM). This depolarizing medium itself inhibited protein synthesis in a manner which was partially mediated by NMDA receptor activation, since it was significantly reversed by the noncompetitive NMDA antagonist, MK-801. Furthermore, the effect of glutamate under depolarizing conditions was also significantly decreased by MK-801, suggesting that glutamate was acting at NMDA receptors. Thus, depolarization appears to enhance the sensitivity of neurons to toxic NMDA receptor activation by glutamate. Under conditions that mimic ischemia, hypoxia plus hypoglycemia, a similar protective effect of NMDA receptor antagonists was observed. Depolarization and ischemia both appeared to attenuate the neurotoxicity of non-NMDA receptor agonists. It appears that under conditions of normal glucose and oxygen, high concentrations of bath applied glutamate inhibit protein synthesis at sites other than the NMDA receptor. However, when the Na+ gradient is decreased, as occurs during ischemia, glutamate's NMDA effects predominate. These findings suggest that ionic shifts may play a central role in permitting NMDA receptor-mediated ischemic neuronal damage.
Collapse
Affiliation(s)
- J J Vornov
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | | |
Collapse
|
34
|
Choi DW. Excitotoxicity on Cultured Cortical Neurons. RESEARCH AND PERSPECTIVES IN NEUROSCIENCES 1991. [DOI: 10.1007/978-3-642-84526-0_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
35
|
Fukuda T, Adachi E, Kawashima S, Yoshiya I, Hashimoto PH. Immunohistochemical distribution of calcium-activated neutral proteinases and endogenous CANP inhibitor in the rabbit hippocampus. J Comp Neurol 1990; 302:100-9. [PMID: 2086609 DOI: 10.1002/cne.903020108] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Intracellular accumulation of Ca2+ after brain ischemia is regarded as one of the principal causes of neuronal death, but details of the intracellular events occurring after Ca2+ accumulation have not yet been described. We propose that a calcium-activated neutral proteinase which can degrade neuronal cytoskeletal proteins might link Ca2+ accumulation and irreversible injury of the neuronal intracellular structure. First, therefore, we examined the distribution of calcium-activated neutral proteinase in normal brains. Immunohistochemical distribution of calcium-activated neutral proteinases (CANP) with high and low sensitivity to Ca2+ (muCANP and mCANP) and of endogenous CANP inhibitor was investigated in the dorsal hippocampus of the rabbit. muCANP-immunoreactivity was detected in almost all of the pyramidal cells and granule cells and in some other neurons. A full-length staining from perikarya to dendrites was shown in muCANP-positive neurons. mCANP-immunoreactivity was found mainly in four kinds of hippocampal interneurons: 1) basket cells in the stratum oriens of Ammon's horn, 2) pyramidal basket cells at the boundary of pyramidal cell layer and stratum oriens, 3) polymorphic cells in the hilar region of dentate gyrus, and 4) pyramidal or fusiform basket cells at the inner boundary of the granule cell layer and the hilar region. The distribution of these four kinds of neurons was similar to that of parvalbumin-containing GABAergic neurons. CANP inhibitor immunoreactivity was confined to pyramidal cells in the CA3-CA3c region and some hilar neurons.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- T Fukuda
- Department of Anatomy, Osaka University Medical School, Japan
| | | | | | | | | |
Collapse
|