1
|
Signaling Pathway of Taurine-Induced Upregulation of TXNIP. Metabolites 2022; 12:metabo12070636. [PMID: 35888758 PMCID: PMC9317136 DOI: 10.3390/metabo12070636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Taurine, a sulfur-containing β-amino acid, is present at high concentrations in mammalian tissues and plays an important role in several essential biological processes. However, the genetic mechanisms involved in these physiological processes associated with taurine remain unclear. In this study, we investigated the regulatory mechanism underlying the taurine-induced transcriptional enhancement of the thioredoxin-interacting protein (TXNIP). The results showed that taurine significantly increased the luciferase activity of the human TXNIP promoter. Further, deletion analysis of the TXNIP promoter showed that taurine induced luciferase activity only in the TXNIP promoter region (+200 to +218). Furthermore, by employing a bioinformatic analysis using the TRANSFAC database, we focused on Tst-1 and Ets-1 as candidates involved in taurine-induced transcription and found that the mutation in the Ets-1 sequence did not enhance transcriptional activity by taurine. Additionally, chromatin immunoprecipitation assays indicated that the binding of Ets-1 to the TXNIP promoter region was enhanced by taurine. Taurine also increased the levels of phosphorylated Ets-1, indicating activation of Ets-1 pathway by taurine. Moreover, an ERK cascade inhibitor significantly suppressed the taurine-induced increase in TXNIP mRNA levels and transcriptional enhancement of TXNIP. These results suggest that taurine enhances TXNIP expression by activating transcription factor Ets-1 via the ERK cascade.
Collapse
|
2
|
Satsu H, Fukumura M, Watari K. Regulation of CXCR4 Expression by Taurine in Macrophage-Like Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:41-49. [DOI: 10.1007/978-3-030-93337-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
3
|
Zhang W, Lan Z, Li K, Liu C, Jiang P, Lu W. Inhibitory role of taurine in the caudal neurosecretory Dahlgren cells of the olive flounder, Paralichthys olivaceus. Gen Comp Endocrinol 2020; 299:113613. [PMID: 32950586 DOI: 10.1016/j.ygcen.2020.113613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 01/19/2023]
Abstract
Taurine plays role in neural development and physiological functions such as endocrine regulation in the central nervous system (CNS), and it is one of the most abundant free amino acid there. We investigated its potential effect as a neurotransmitter in the group of neuroendocrine Dahlgren cells at flounder Paralichthys olivaceus caudal neurosecretory system (CNSS). The application of taurine in vitro led to a reduction in electrical activity of Dahlgren cells, followed by a rise in the number of silent cells, at the same time the frequency of all three activity patterns (tonic, phasic, bursting) in Dahlgren cells was reduced. Both strychnine (a glycine receptor antagonist) and bicuculline (a GABAA receptor antagonist) can block the response to taurine separately. Transcriptome sequencing analysis showed the existence of glycine receptor (GlyR) and GABAA receptor (GABAAR) in the flounder CNSS, and the GlyR, GABAAR, and Cl- channel mRNA expression were significantly raised after taurine superfusion according to quantitative RT-PCR results. These data indicate that taurine may mediate Dahlgren cell population of CNSS activity in vivo through GlyR and GABAAR, thereby, regulating stress-response.
Collapse
Affiliation(s)
- Wei Zhang
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Zhaohui Lan
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Kunyu Li
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Cheng Liu
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Pengxin Jiang
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Weiqun Lu
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China.
| |
Collapse
|
4
|
Jakaria M, Azam S, Haque ME, Jo SH, Uddin MS, Kim IS, Choi DK. Taurine and its analogs in neurological disorders: Focus on therapeutic potential and molecular mechanisms. Redox Biol 2019; 24:101223. [PMID: 31141786 PMCID: PMC6536745 DOI: 10.1016/j.redox.2019.101223] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/21/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Taurine is a sulfur-containing amino acid and known as semi-essential in mammals and is produced chiefly by the liver and kidney. It presents in different organs, including retina, brain, heart and placenta and demonstrates extensive physiological activities within the body. In the several disease models, it attenuates inflammation- and oxidative stress-mediated injuries. Taurine also modulates ER stress, Ca2+ homeostasis and neuronal activity at the molecular level as part of its broader roles. Different cellular processes such as energy metabolism, gene expression, osmosis and quality control of protein are regulated by taurine. In addition, taurine displays potential ameliorating effects against different neurological disorders such as neurodegenerative diseases, stroke, epilepsy and diabetic neuropathy and protects against injuries and toxicities of the nervous system. Several findings demonstrate its therapeutic role against neurodevelopmental disorders, including Angelman syndrome, Fragile X syndrome, sleep-wake disorders, neural tube defects and attention-deficit hyperactivity disorder. Considering current biopharmaceutical limitations, developing novel delivery approaches and new derivatives and precursors of taurine may be an attractive option for treating neurological disorders. Herein, we present an overview on the therapeutic potential of taurine against neurological disorders and highlight clinical studies and its molecular mechanistic roles. This article also addresses the neuropharmacological potential of taurine analogs.
Collapse
Affiliation(s)
- Md Jakaria
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Shofiul Azam
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Md Ezazul Haque
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Song-Hee Jo
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - In-Su Kim
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea; Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, and Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences and Integrated Bioscience, Graduate School, Konkuk University, Chungju, South Korea; Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Sciences, and Research Institute of Inflammatory Diseases (RID), Konkuk University, Chungju, South Korea.
| |
Collapse
|
5
|
Rescue of the Functional Alterations of Motor Cortical Circuits in Arginase Deficiency by Neonatal Gene Therapy. J Neurosci 2017; 36:6680-90. [PMID: 27335400 DOI: 10.1523/jneurosci.0897-16.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/11/2016] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED Arginase 1 deficiency is a urea cycle disorder associated with hyperargininemia, spastic diplegia, loss of ambulation, intellectual disability, and seizures. To gain insight on how loss of arginase expression affects the excitability and synaptic connectivity of the cortical neurons in the developing brain, we used anatomical, ultrastructural, and electrophysiological techniques to determine how single-copy and double-copy arginase deletion affects cortical circuits in mice. We find that the loss of arginase 1 expression results in decreased dendritic complexity, decreased excitatory and inhibitory synapse numbers, decreased intrinsic excitability, and altered synaptic transmission in layer 5 motor cortical neurons. Hepatic arginase 1 gene therapy using adeno-associated virus rescued nearly all these abnormalities when administered to neonatal homozygous knock-out animals. Therefore, gene therapeutic strategies can reverse physiological and anatomical markers of arginase 1 deficiency and therefore may be of therapeutic benefit for the neurological disabilities in this syndrome. SIGNIFICANCE STATEMENT These studies are one of the few investigations to try to understand the underlying neurological dysfunction that occurs in urea cycle disorders and the only to examine arginase deficiency. We have demonstrated by multiple modalities that, in murine layer 5 cortical neurons, a gradation of abnormalities exists based on the functional copy number of arginase: intrinsic excitability is altered, there is decreased density in asymmetrical and perisomatic synapses, and analysis of the dendritic complexity is lowest in the homozygous knock-out. With neonatal administration of adeno-associated virus expressing arginase, there is near-total recovery of the abnormalities in neurons and cortical circuits, supporting the concept that neonatal gene therapy may prevent the functional abnormalities that occur in arginase deficiency.
Collapse
|
6
|
Winge I, Teigen K, Fossbakk A, Mahootchi E, Kleppe R, Sköldberg F, Kämpe O, Haavik J. Mammalian CSAD and GADL1 have distinct biochemical properties and patterns of brain expression. Neurochem Int 2015; 90:173-84. [PMID: 26327310 DOI: 10.1016/j.neuint.2015.08.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/30/2015] [Accepted: 08/22/2015] [Indexed: 11/17/2022]
Abstract
Variants in the gene encoding the enzyme glutamic acid decarboxylase like 1 (GADL1) have been associated with response to lithium therapy. Both GADL1 and the related enzyme cysteine sulfinic acid decarboxylase (CSAD) have been proposed to be involved in the pyridoxal-5'-phosphate (PLP)-dependent biosynthesis of taurine. In the present study, we compared the catalytic properties, inhibitor sensitivity and expression profiles of GADL1 and CSAD in brain tissue. In mouse and human brain we observed distinct patterns of expression of the PLP-dependent decarboxylases CSAD, GADL1 and glutamic acid decarboxylase 67 (GAD67). CSAD levels were highest during prenatal and early postnatal development; GADL1 peaked early in prenatal development, while GAD67 increased rapidly after birth. Both CSAD and GADL1 are being expressed in neurons, whereas only CSAD mRNA was detected in astrocytes. Cysteine sulfinic acid was the preferred substrate for both mouse CSAD and GADL1, although both enzymes also decarboxylated cysteic acid and aspartate. In silico screening and molecular docking using the crystal structure of CSAD and in vitro assays led to the discovery of eight new enzyme inhibitors with partial selectivity for either CSAD or GADL1. Lithium had minimal effect on their enzyme activities. In conclusion, taurine biosynthesis in vertebrates involves two structurally related PLP-dependent decarboxylases (CSAD and GADL1) that have partially overlapping catalytic properties but different tissue distribution, indicating divergent physiological roles. Development of selective enzyme inhibitors targeting these enzymes is important to further dissect their (patho)physiological roles.
Collapse
Affiliation(s)
- Ingeborg Winge
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway
| | - Knut Teigen
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway
| | - Agnete Fossbakk
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway
| | - Elaheh Mahootchi
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway
| | - Rune Kleppe
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway
| | - Filip Sköldberg
- Department of Medical Sciences, University Hospital, Uppsala University, Uppsala, Sweden
| | - Olle Kämpe
- Department of Medical Sciences, University Hospital, Uppsala University, Uppsala, Sweden; Centre of Molecular Medicine (CMM L8:01), Dept. of Medicine (Solna), Karolinska Instituttet, Stockholm, Sweden
| | - Jan Haavik
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
7
|
Abstract
Abstract:Taurine appears to exert potent protections against glutamate (Glu)-induced injury to neurons, but the underlying molecular mechanisms are not fully understood. The possibly protected targets consist of the plasma membrane and the mitochondrial as well as endoplasmic reticulum (ER) membranes. Protection may be provided through a variety of effects, including the prevention of membrane depolarization, neuronal excitotoxicity and mitochondrial energy failure, increases in intracellular free calcium ([Ca2+]i), activation of calpain, and reduction of Bcl-2 levels. These activities are likely to be linked spatially and temporally in the neuroprotective functions of taurine. In addition, events that occur downstream of Glu stimulation, including altered enzymatic activities, apoptotic pathways, and necrosis triggered by the increased [Ca2+]i, can be inhibited by taurine. This review discusses the possible molecular mechanisms of taurine against Glu-induced neuronal injury, providing a better understanding of the protective processes, which might be helpful in the development of novel interventional strategies.
Collapse
|
8
|
Ye HB, Wang J, Zhang WT, Shi HB, Yin SK. Taurine attenuates bilirubin-induced neurotoxicity in the auditory system in neonatal guinea pigs. Int J Pediatr Otorhinolaryngol 2013; 77:647-54. [PMID: 23273639 DOI: 10.1016/j.ijporl.2012.11.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 11/17/2012] [Accepted: 11/20/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVES Previous work showed that taurine protects neurons against unconjugated bilirubin (UCB)-induced neurotoxicity by maintaining intracellular calcium homeostasis, membrane integrity, and mitochondrial function, thereby preventing apoptosis from occurring, in primary neuron cultures. In this study, we investigated whether taurine could protect the auditory system against the neurotoxicity associated with hyperbilirubinemia in an in vivo model. METHODS Hyperbilirubinemia was established in neonatal guinea pigs by intraperitoneal injection of UCB. Hearing function was observed in electrocochleograms (ECochGs) and auditory brainstem responses (ABRs) recorded before and 1, 8, 24, and 72 h after UCB injection. For morphological evaluations, animals were sacrificed at 8h post-injection, and the afferent terminals beneath the inner hair cells (IHCs), spiral ganglion neurons (SGNs), and their fibers were examined. RESULTS It was found that UCB injection significantly increased latencies and inter-wave intervals, and thresholds of ABR and compound action potentials, and caused marked damage to type I SGNs, their axons, and terminals to cochlear IHCs. When baby guinea pigs were pretreated with taurine for 5 consecutive days and then injected with bilirubin, electrophysiological abnormalities and morphological damage were attenuated significantly in both the peripheral and central auditory system. CONCLUSIONS From these observations, it was concluded that taurine limited bilirubin-induced neural damage in the auditory system. These findings may contribute to the development of taurine as a broad-spectrum agent for preventing and/or treating hearing loss in neonatal jaundice.
Collapse
Affiliation(s)
- Hai-Bo Ye
- Department of Otorhinolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiaotong University, 600 Yishan Road, Shanghai 200233, China
| | | | | | | | | |
Collapse
|
9
|
Taurine and Its Neuroprotective Role. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 775:19-27. [DOI: 10.1007/978-1-4614-6130-2_2] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Oja SS, Saransaari P. Regulation of Taurine Release in the Hippocampus of Developing and Adult Mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 775:135-43. [DOI: 10.1007/978-1-4614-6130-2_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
11
|
Advances in drug design based on the amino Acid approach: taurine analogues for the treatment of CNS diseases. Pharmaceuticals (Basel) 2012; 5:1128-46. [PMID: 24281261 PMCID: PMC3816653 DOI: 10.3390/ph5101128] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/24/2012] [Accepted: 10/15/2012] [Indexed: 11/17/2022] Open
Abstract
Amino acids are well known to be an important class of compounds for the maintenance of body homeostasis and their deficit, even for the polar neuroactive aminoacids, can be controlled by supplementation. However, for the amino acid taurine (2-aminoethanesulfonic acid) this is not true. Due its special physicochemical properties, taurine is unable to cross the blood-brain barrier. In addition of injured taurine transport systems under pathological conditions, CNS supplementation of taurine is almost null. Taurine is a potent antioxidant and anti-inflammatory semi-essential amino acid extensively involved in neurological activities, acting as neurotrophic factor, binding to GABA A/glycine receptors and blocking the excitotoxicity glutamate-induced pathway leading to be a neuroprotective effect and neuromodulation. Taurine deficits have been implicated in several CNS diseases, such as Alzheimer’s, Parkinson’s, epilepsy and in the damage of retinal neurons. This review describes the CNS physiological functions of taurine and the development of new derivatives based on its structure useful in CNS disease treatment.
Collapse
|
12
|
Interaction between taurine and GABA(A)/glycine receptors in neurons of the rat anteroventral cochlear nucleus. Brain Res 2012; 1472:1-10. [PMID: 22796293 DOI: 10.1016/j.brainres.2012.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 06/30/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
Abstract
Taurine, one of the most abundant endogenous amino acids in the mammalian central nervous system (CNS), is involved in neural development and many physiological functions. In this study, the interaction between taurine and GABA(A)/glycine receptors was investigated in young rat (P13-P15) anteroventral cochlear nucleus (AVCN) neurons using the whole-cell patch-clamp method. We found that taurine at low (0.1mM) and high (1mM) concentrations activated both GABA(A) and glycine receptors, but not AMPA and NMDA receptors. The reversal potentials of taurine-, GABA- or glycine-evoked currents were close to the expected chloride equilibrium potential, indicating that receptors activated by these agonists were mediating chloride conductance. Moreover, our results showed that the currents activated by co-application of GABA and glycine were cross-inhibitive. Sequential application of GABA and glycine or vice versa also reduced the glycine or GABA evoked currents. There was no cross-inhibition when taurine and GABA or taurine and glycine were applied simultaneously, but the response was larger than that evoked by GABA or glycine alone. These results suggest that taurine can serve as a neuromodulator to strengthen GABAergic and glycinergic neurotransmission in the rat AVCN.
Collapse
|
13
|
Abstract
Taurine demonstrates multiple cellular functions including a central role as a neurotransmitter, as a trophic factor in CNS development, in maintaining the structural integrity of the membrane, in regulating calcium transport and homeostasis, as an osmolyte, as a neuromodulator and as a neuroprotectant. The neurotransmitter properties of taurine are illustrated by its ability to elicit neuronal hyperpolarization, the presence of specific taurine synthesizing enzyme and receptors in the CNS and the presence of a taurine transporter system. Taurine exerts its neuroprotective functions against the glutamate induced excitotoxicity by reducing the glutamate-induced increase of intracellular calcium level, by shifting the ratio of Bcl-2 and Bad ratio in favor of cell survival and by reducing the ER stress. The presence of metabotropic taurine receptors which are negatively coupled to phospholipase C (PLC) signaling pathway through inhibitory G proteins is proposed, and the evidence supporting this notion is also presented.
Collapse
Affiliation(s)
- Jang-Yen Wu
- Charles E. Schmidt College of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Howard Prentice
- Charles E. Schmidt College of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
14
|
Taurine protects against bilirubin-induced neurotoxicity in vitro. Brain Res 2010; 1320:159-67. [DOI: 10.1016/j.brainres.2010.01.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 02/03/2023]
|
15
|
Park EJ, Choi IS, Cho JH, Nakamura M, Lee JJ, Lee MG, Choi BJ, Moorhouse AJ, Jang IS. Zinc modulation of glycine receptors in acutely isolated rat CA3 neurons. Life Sci 2008; 83:149-54. [DOI: 10.1016/j.lfs.2008.04.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 04/25/2008] [Accepted: 04/25/2008] [Indexed: 10/21/2022]
|
16
|
Abstract
Our knowledge of glycine receptor (GlyR) regulation of excitation has advanced significantly in recent years. GlyRs are widespread in the CNS, are heterogeneous, and undergo developmental changes. Activation of GlyRs of immature neurons induces outflow of Cl( - ), membrane depolarization, neuronal excitation, calcium influx, and transmitter release, in contrast to the inhibitory effects these receptors have in mature neurons. Thus, GlyRs are important for neuronal excitability in both the developing and the mature CNS. This chapter is an overview of selective studies on the newly discovered roles of GlyRs in regulating neuronal excitation, and inhibition, particularly in the upper brain areas.
Collapse
Affiliation(s)
- Jiang-Hong Ye
- Department of Anesthesiology, UMDNJ, New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
17
|
Janáky R, Shaw CA, Oja SS, Saransaari P. Taurine release in developing mouse hippocampus is modulated by glutathione and glutathione derivatives. Amino Acids 2007; 34:75-80. [PMID: 17701096 PMCID: PMC2814815 DOI: 10.1007/s00726-007-0587-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Accepted: 07/08/2007] [Indexed: 11/24/2022]
Abstract
Glutathione (reduced form GSH and oxidized form GSSG) constitutes an important defense against oxidative stress in the brain, and taurine is an inhibitory neuromodulator particularly in the developing brain. The effects of GSH and GSSG and glycylglycine, gamma-glutamylcysteine, cysteinylglycine, glycine and cysteine on the release of [(3)H]taurine evoked by K+-depolarization or the ionotropic glutamate receptor agonists glutamate, kainate, 2-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) and N-methyl-D-aspartate (NMDA) were now studied in slices from the hippocampi from 7-day-old mouse pups in a perfusion system. All stimulatory agents (50 mM K(+), 1 mM glutamate, 0.1 mM kainate, 0.1 mM AMPA and 0.1 mM NMDA) evoked taurine release in a receptor-mediated manner. Both GSH and GSSG significantly inhibited the release evoked by 50 mM K+. The release induced by AMPA and glutamate was also inhibited, while the kainate-evoked release was significantly activated by both GSH and GSSG. The NMDA-evoked release proved the most sensitive to modulation: L-Cysteine and glycine enhanced the release in a concentration-dependent manner, whereas GSH and GSSG were inhibitory at low (0.1 mM) but not at higher (1 or 10 mM) concentrations. The release evoked by 0.1 mM AMPA was inhibited by gamma-glutamylcysteine and cysteinylglycine, whereas glycylglycine had no effect. The 0.1 mM NMDA-evoked release was inhibited by glycylglycine and gamma-glutamylcysteine. In turn, cysteinylglycine inhibited the NMDA-evoked release at 0.1 mM, but was inactive at 1 mM. Glutathione exhibited both enhancing and attenuating effects on taurine release, depending on the glutathione concentration and on the agonist used. Both glutathione and taurine act as endogenous neuroprotective effectors during early postnatal life.
Collapse
Affiliation(s)
- R Janáky
- Brain Research Center, University of Tampere Medical School, Tampere, Finland
| | | | | | | |
Collapse
|
18
|
Anderzhanova E, Saransaari P, Oja SS. Neuroprotective mechanisms of taurine in vivo. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 583:377-87. [PMID: 17153623 DOI: 10.1007/978-0-387-33504-9_42] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
|
19
|
Chepkova AN, Sergeeva OA, Haas HL. Taurine rescues hippocampal long-term potentiation from ammonia-induced impairment. Neurobiol Dis 2006; 23:512-21. [PMID: 16766203 DOI: 10.1016/j.nbd.2006.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 04/11/2006] [Accepted: 04/12/2006] [Indexed: 11/19/2022] Open
Abstract
Hyperammonemia, a major pathophysiological factor in hepatic encephalopathy, impairs long-term potentiation (LTP) of synaptic transmission, a cellular model of learning and memory, in the hippocampus. We have now studied the protective action of taurine on this paradigm by analyzing LTP characteristics in mouse hippocampal slices treated with ammonium chloride (1 mM) in the presence of taurine (1 mM), an ubiquitous osmolyte, antioxidant, and neuromodulator, as well as other substances with such properties. Ammonia-treated slices displayed a significant impairment of LTP maintenance. Taurine and the mitochondrial enhancer l-carnitine, but not the antioxidants (ascorbate, carnosine, and the novel compound GVS-111) or the osmolyte betaine prevented this impairment. The protective effect of taurine was preserved under the blockade of inhibitory GABA(A) and glycine receptors. It is suggested that taurine may rescue the mechanisms of hippocampal synaptic plasticity by improving mitochondrial function under hyperammonemic conditions.
Collapse
Affiliation(s)
- Aisa N Chepkova
- Department of Neurophysiology, Heinrich-Heine University, POB 101007, D-40001 Düsseldorf, Germany
| | | | | |
Collapse
|
20
|
Wang F, Xiao C, Ye JH. Taurine activates excitatory non-synaptic glycine receptors on dopamine neurones in ventral tegmental area of young rats. J Physiol 2005; 565:503-16. [PMID: 15817633 PMCID: PMC1464534 DOI: 10.1113/jphysiol.2005.085423] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The physiological and pharmacological properties of taurine-induced responses were investigated in dopaminergic (DA) neurones from the ventral tegmental area (VTA) of young rats aged 1-13 postnatal days, either in acute brain slices or acutely dissociated neurones. When whole-cell responses were recorded from current-clamped neurones using the gramicidin-perforated technique, the application of taurine (0.01-30 mm) accelerated firings and induced membrane depolarization. In voltage-clamped neurones, taurine induced a current which was antagonized by strychnine and by picrotoxin, but not by bicuculline. In addition, taurine-induced current showed complete cross-desensitization with glycine-activated currents but not with gamma-aminobutyric acid (GABA)-activated currents. Thus, taurine is a full agonist of the glycine receptors (GlyRs) in the VTA. Further studies found that taurine acted mainly on non-synaptic GlyRs. The application of 20 microm bicuculline abolished the spontaneous inhibitory post-synaptic currents (IPSCs) in 40/45 neurones, and 93% of the evoked IPSCs. The addition of 1 microm strychnine completely eliminated the remaining IPSCs. These results suggest that GABAergic IPSCs predominate, and that functional glycinergic synapses are present in a subset of the VTA neurones. The application of 1 mum strychnine alone induced an outward current, suggesting that these neurones were exposed to tonically released taurine/glycine. In conclusion, by activating non-synaptic GlyRs, taurine may act as an excitatory extra-synaptic neurotransmitter in the VTA during early development.
Collapse
Affiliation(s)
- Fushun Wang
- New Jersey Medical School (UMDNJ), 185 South Orange Avenue, Newark, NJ 07103-2714, USA
| | | | | |
Collapse
|
21
|
Wu H, Jin Y, Wei J, Jin H, Sha D, Wu JY. Mode of action of taurine as a neuroprotector. Brain Res 2005; 1038:123-31. [PMID: 15757628 DOI: 10.1016/j.brainres.2005.01.058] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2004] [Revised: 12/23/2004] [Accepted: 01/01/2005] [Indexed: 10/25/2022]
Abstract
Previously, it has been shown that taurine exerts its protective function against glutamate-induced neuronal excitotoxicity through its action in reducing glutamate-induced elevation of intracellular free calcium, [Ca2+]i. Here, we report the mechanism underlying the effect of taurine in reducing [Ca2+]i. We found that taurine inhibited glutamate-induced calcium influx through L-, P/Q-, N-type voltage-gated calcium channels (VGCCs) and NMDA receptor calcium channel. Surprisingly, taurine had no effect on calcium influx through NMDA receptor calcium channel when cultured neurons were treated with NMDA in Mg2+-free medium. Since taurine was found to prevent glutamate-induced membrane depolarization, we propose that taurine protects neurons against glutamate excitotoxicity by preventing glutamate-induced membrane depolarization, probably through its effect in opening of chloride channels and, therefore, preventing the glutamate-induced increase in calcium influx and other downstream events.
Collapse
Affiliation(s)
- Heng Wu
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | | | | | | | | | | |
Collapse
|
22
|
Belluzzi O, Puopolo M, Benedusi M, Kratskin I. Selective neuroinhibitory effects of taurine in slices of rat main olfactory bulb. Neuroscience 2004; 124:929-44. [PMID: 15026133 DOI: 10.1016/j.neuroscience.2003.12.032] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2003] [Indexed: 01/20/2023]
Abstract
Taurine is abundant in the main olfactory bulb, exceeding glutamate and GABA in concentration. In whole-cell patch-clamp recordings in rat olfactory bulb slices, taurine inhibited principal neurons, mitral and tufted cells. In these cells, taurine decreased the input resistance and caused a shift of the membrane potential toward the chloride equilibrium potential. The taurine actions were sustained under the blockade of transmitter release and were reversible and dose-dependent. At a concentration of 5 mM, typically used in this study, taurine showed 90% of its maximal effect. GABA(A) antagonists, bicuculline and picrotoxin, blocked the taurine actions, whereas the glycine receptor antagonist strychnine and GABA(B) antagonists, CGP 55845A and CGP 35348, were ineffective. These findings are consistent with taurine directly activating GABA(A) receptors and inducing chloride conductance. Taurine had no effect on periglomerular and granule interneurons. The subunit composition of GABA(A) receptors in these cells, differing from those in mitral and tufted cells, may account for taurine insensitivity of the interneurons. Taurine suppressed olfactory nerve-evoked monosynaptic responses of mitral and tufted cells while chloride conductance was blocked. This action was mimicked by the GABA(B) agonist baclofen and abolished by CGP 55845A; CGP 35348, which primarily blocks postsynaptic GABA(B) receptors, was ineffective. The taurine effect most likely was due to GABA(B) receptor-mediated inhibition of presynaptic glutamate release. Neither taurine nor baclofen affected responses of periglomerular cells. The lack of a baclofen effect implies that functional GABA(B) receptors are absent from olfactory nerve terminals that contact periglomerular cells. These results indicate that taurine decreases the excitability of mitral and tufted cells and their responses to olfactory nerve stimulation without influencing periglomerular and granule cells. Selective effects of taurine in the olfactory bulb may represent a physiologic mechanism that is involved in the inhibitory shaping of the activation pattern of principal neurons.
Collapse
Affiliation(s)
- O Belluzzi
- Department of Biology, Section of Physiology and Biophysics, Center of Neurosciences, University of Ferrara, 46 Via Borsari, 44100 Ferrara, Italy.
| | | | | | | |
Collapse
|
23
|
Satoh H. Elecropharmacology of taurine on the hyperpolarization-activated inward current and the sustained inward current in spontaneously beating rat sino-atrial nodal cells. J Pharmacol Sci 2003; 91:229-38. [PMID: 12686746 DOI: 10.1254/jphs.91.229] [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] [Indexed: 10/27/2022] Open
Abstract
Modulation by taurine of the pacemaking activity and the underlying ionic currents, especially a hyperpolarization-activated inward current (I(f)) and a sustained inward current (I(ST)), in rat sino-atrial (SA) nodal cells was investigated at different pCa levels using a patch-clamp technique. Increasing pCa levels from 10 to 6 stimulated the spontaneous activity and simultaneously increased the I(f). Application of taurine depressed more strongly the spontaneous activity at higher pCa levels. At all pCa levels, however, taurine (20 mM) increased the I(f) by 60.1 +/- 1.7% (n = 8, P<0.001) at pCa 10 and by 48.0 +/- 1.4% (n = 8, P<0.01) at pCa 7. At pCa 7, taurine (10 and 20 mM) decreased the sustained inward current (I(ST)) by 13.3 +/- 1.1% (n = 5, P<0.05) and by 38.1 +/- 2.4% (n = 5, P<0.01), respectively. Taurine (20 mM) inhibited the L-type Ca(2+) current (I(CaL)) by 35.8 +/- 2.5% (n = 8, P<0.01), whereas taurine enhanced the T-type Ca(2+) current (I(CaT)) by 29.3 +/- 2.9% (n = 8, P<0.05). Also, taurine at pCa 7 decreased the delayed rectifier K(+) current; taurine at 20 mM inhibited the rapidly activated K(+) current (I(Kr)) by 55.6 +/- 3.3% (n = 6, P<0.001), but not the slowly activated K(+) current (I(Ks)). Taurine often elicited dysrhythmias, dependent on taurine's concentrations and pCa levels. These results indicate that taurine causes a negative chronotropic effect due to the inhibitions of the pacemaking ionic currents such as I(CaL), I(Kr) and I(ST), and suggest that the I(f) and I(CaT) currents make a minor contribution to pacemaking activity in rat SA nodal cells.
Collapse
Affiliation(s)
- Hiroyasu Satoh
- Department of Pharmacology, Division of Molecular and Cellular Biology, Nara Medical University, Japan.
| |
Collapse
|
24
|
Yu K, Ge SY, Ruan DY. Fe2+ decreases the taurine-induced Cl- current in acutely dissociated rat hippocampal neurons. Brain Res 2003; 960:25-35. [PMID: 12505654 DOI: 10.1016/s0006-8993(02)03736-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effects of ferrous ions (Fe(2+)) on taurine-induced Cl(-) current (I(tau)) recorded from single neurons, which was freshly isolated from the rat hippocampal CA1 area, were studied with conventional whole-cell recording under voltage-clamp conditions. Using standard pharmacological approaches, we found that the currents gated by concentrations of taurine (<or=10 mM), which existed in about 90% of the hippocampal neurons tested, were predominantly mediated by strychnine-sensitive glycine receptors. When co-applied with taurine, Fe(2+) effectively depressed I(tau) in a concentration-dependent manner, with an IC(50) of 3.76 mM and Hill coefficient of 1.01, while preincubation with 1 mM Fe(2+) alone did not affect the following membrane currents elicited by taurine. The result suggests that resting taurine-gated channels are insensitive to Fe(2+). Since internal cell dialysis with 3 mM Fe(2+) failed to modify I(tau), it was deduced that the site of action of Fe(2+) is extracellular. Furthermore, the Lineweaver-Burke double reciprocal plot of normalized response to taurine against the concentration of taurine illustrated that the depression of I(tau) was noncompetitive, therefore Fe(2+) may act on the glycine receptor-chloride ionophore complex at a site distinct from where taurine binds. Various concentrations of Fe(2+) ranging from 0.1 to 20 mM depressed I(tau) and this extracellular depression was independent of membrane voltage. These results indicate that Fe(2+) decreases I(tau) in acutely dissociated rat hippocampal neurons and the inhibition of glycine receptors by Fe(2+) might be one possible approach through which Fe(2+) induces seizures.
Collapse
Affiliation(s)
- Kuai Yu
- School of Life Science, University of Science and Technology of China, Hefei Anhui, 230027 PR China
| | | | | |
Collapse
|
25
|
Quertemont E, Devitgh A, De Witte P. Systemic osmotic manipulations modulate ethanol-induced taurine release: a brain microdialysis study. Alcohol 2003; 29:11-9. [PMID: 12657372 DOI: 10.1016/s0741-8329(02)00324-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In recent microdialysis studies, increased extracellular concentrations of taurine after high ethanol dose administration were identified in various rat brain regions. The mechanisms by which ethanol caused these increases in extracellular taurine concentration remained unclear but could be related to ethanol-induced cell swelling. The aim of the current study was to investigate whether changes in the body osmotic state modulate the effects of ethanol on brain extracellular taurine concentrations. In several groups of rats, brain hypoosmotic or hyperosmotic states were superimposed on acute ethanol (2.0-g/kg) injections, and extracellular taurine concentrations within the nucleus accumbens were assessed by using an intracerebral microdialysis procedure. A hypoosmotic state was obtained by systemic administration of water while hyperosmotic states were induced by intraperitoneal injections of hypertonic saline solutions (1.8% or 3.6% saline). In isoosmotic conditions, ethanol induced an immediate and significant increase in taurine microdialysate content, confirming results of previous studies. However, the effects of ethanol on taurine concentrations were modulated by osmotic manipulations. Hypoosmotic conditions significantly potentiated ethanol-induced taurine release. In contrast, ethanol-induced increases in extracellular taurine levels were attenuated by 1.8% saline injection and totally prevented by 3.6% saline administration. These results strongly argue in favor of a primary role of osmoregulation in ethanol-induced taurine release. Ethanol-induced cell swelling probably activates volume-sensitive channels, and taurine passively diffuses outside the cells along its concentration gradient.
Collapse
Affiliation(s)
- Etienne Quertemont
- Biologie du Comportement, Université catholique de Louvain, Place Croix du Sud 1, 1348 Louvain-la-Neuve, Belgium.
| | | | | |
Collapse
|
26
|
Saransaari P, Oja SS. Mechanisms of adenosine release in the developing and adult mouse hippocampus. Neurochem Res 2002; 27:911-8. [PMID: 12396102 DOI: 10.1023/a:1020343631833] [Citation(s) in RCA: 7] [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
Adenosine is a neuromodulator known to inhibit the synaptic release of neurotransmitters, e.g., glutamate, and to hyperpolarize postsynaptic neurons. The release of adenosine is markedly enhanced under ischemic conditions. It may then act as an endogenous neuroprotectant against cerebral ischemia and excitotoxic neuronal damage. The mechanisms by which adenosine is released from nervous tissue are not fully known, particularly in the immature brain. We now characterized the release of [3H]adenosine from hippocampal slices from developing (7-day-old) and adult (3-month-old) mice using a superfusion system. The properties of the release differed only partially in the immature and mature hippocampus. The K(+)-evoked release was Ca2+ and Na+ dependent. Anion channels were also involved. Ionotropic glutamate receptor agonists potentiated the release in a receptor-mediated manner. Activation of metabotropic glutamate receptors enhanced the release in developing mice, with group II receptors alone being effective. The evoked adenosine release apparently provides neuroprotective effects against excitotoxicity under cell-damaging conditions. Taurine had no effect on adenosine release in adult mice, but depressed the release concentration dependently in the immature hippocampus.
Collapse
Affiliation(s)
- Pirjo Saransaari
- Tampere Brain Research Center, Medical School, University of Tampere, Finland.
| | | |
Collapse
|
27
|
Chattipakorn SC, McMahon LL. Pharmacological characterization of glycine-gated chloride currents recorded in rat hippocampal slices. J Neurophysiol 2002; 87:1515-25. [PMID: 11877523 DOI: 10.1152/jn.00365.2001] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An inhibitory role for strychnine-sensitive glycine-gated chloride channels (GlyRs) in mature hippocampus has been overlooked, largely due to the misconception that GlyR expression ceases early during development and to few functional studies demonstrating their presence. As a result, little is known regarding the physiological and pharmacological properties of native GlyRs expressed by hippocampal neurons. In this study, we used pharmacological tools and whole cell patch-clamp recordings of CA1 pyramidal cells and interneurons in acutely prepared hippocampal slices from 3- to 4-wk old rats to characterize these understudied receptors. We show that glycine application to recorded pyramidal cells and interneurons elicited strychnine-sensitive chloride-mediated currents (I(gly)) that did not completely desensitize in the continued presence of agonist but reached a steady state at 45-60% of the peak amplitude. Additionally, the inhibitory amino acid, taurine, which has been shown to activate GlyRs in other systems, activated GlyRs expressed by both pyramidal cells and interneurons, although with much less potency than glycine, having an EC(50) 10-fold higher. To examine the potential subunit composition of hippocampal GlyRs, we tested the effect of the GABA(A) receptor antagonist, picrotoxin, on I(gly) recorded from both cell types. At low micromolar concentrations of picrotoxin (< or =100 microM), which selectively block alpha homomeric GlyRs, I(gly) was partially attenuated in both cell types, indicating that alpha homomeric receptors are expressed by pyramidal cells and interneurons. At picrotoxin concentrations < or =1 mM, approximately 10-20% of the whole cell current remained, suggesting that alphabeta heteromeric GlyRs are also expressed because this subtype of GlyR is relatively resistant to picrotoxin antagonism. Finally, we examined whether hippocampal GlyRs are modulated by zinc. Consistent with previous reports in other preparations, zinc elicited a bidirectional modulation of GlyRs, with physiological zinc concentrations (1-100 microM) increasing whole cell currents and concentrations >100 microM depressing them. Furthermore, the same concentration of zinc that potentiates I(gly) suppressed currents mediated by the N-methyl-D-aspartate subtype of the glutamate receptor. Thus we provide a pharmacological characterization of native GlyRs expressed by both major neuron types in hippocampus and show that these receptors can be activated by taurine, an amino acid that is highly concentrated in hippocampus. Furthermore, our data suggest that at least two GlyR subtypes are present in hippocampus and that GlyR-mediated currents can be potentiated by zinc at concentrations that suppress glutamate-mediated excitability.
Collapse
Affiliation(s)
- Siriporn C Chattipakorn
- Department of Physiology and Biophysics, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, USA
| | | |
Collapse
|
28
|
Kondziella D, Lüdemann W, Brinker T, Sletvold O, Sonnewald U. Alterations in brain metabolism, CNS morphology and CSF dynamics in adult rats with kaolin-induced hydrocephalus. Brain Res 2002; 927:35-41. [PMID: 11814430 DOI: 10.1016/s0006-8993(01)03320-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study describes the biochemical changes, morphological development and the cerebrospinal fluid dynamics of the kaolin-induced hydrocephalus in the adult rat. Two, 4 and 6 weeks after microsurgical kaolin instillation into the rat cisterna magna the basal intracranial pressure and the cerebrospinal fluid outflow resistance were measured. To determine possible biochemical changes in the rat cerebrum, brain stem and cerebellum the concentrations of glutamine, glutamate, glutathione, aspartate, GABA, alanine and taurine were measured by high pressure liquid chromatography. In addition, ventriculomegaly and syringomyelia were assessed, measuring the lateral ventricles and central canals by means of an image-processing computer program. It could be shown that the acute phase of kaolin-induced hydrocephalus in the first 4 weeks is characterised by a high basal intracranial pressure, a considerably increased CSF outflow resistance and a rise in brain water content in the fourth week. The changes in the concentrations of amino acids were moderate. Glutamine was increased and taurine was decreased in the cerebrum and alanine was increased in the brain stem. The chronic phase, however, is defined by normal basal pressure, declining outflow resistance, progression of ventriculomegaly and distinct changes in the biochemical parameters such as a remarkable decrease of glutamate, glutamine and taurine in the cerebellum, a decrease of taurine and alanine plus an increase in glutamine in the cerebrum and an increase of alanine in the brain stem. Moreover, cerebral metabolism in the adult rat seems to be more resistant to the effects of hydrocephalus than metabolism in neonatal and infantile rats.
Collapse
Affiliation(s)
- Daniel Kondziella
- Department of Clinical Neuroscience, Faculty of Medicine, Olav Kyrresgt. 3, N-7489 Trondheim, Norway
| | | | | | | | | |
Collapse
|
29
|
Foos TM, Wu JY. The role of taurine in the central nervous system and the modulation of intracellular calcium homeostasis. Neurochem Res 2002; 27:21-6. [PMID: 11926272 DOI: 10.1023/a:1014890219513] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of taurine in the mammalian nervous system are numerous and varied. There has been great difficulty in determining the specific targets of taurine action. The authors present a review of accepted taurine action and highlight recent discoveries regarding taurine and calcium homeostasis in neurons. In general there is a consensus that taurine is a powerful agent in regulating and reducing the intracellular calcium levels in neurons. After prolonged L-glutamate stimulation, neurons lose the ability to effectively regulate intracellular calcium. This condition can lead to acute swelling and lysis of the cell, or culminate in apoptosis. Under these conditions, significant amounts of taurine (mM range) are released from the excited neuron. This extracellular taurine acts to slow the influx of calcium into the cytosol through both transmembrane ion transporters and intracellular storage pools. Two specific targets of taurine action are discussed: Na(+)-Ca2+ exchangers, and metabotropic receptors mediating phospholipase-C.
Collapse
Affiliation(s)
- Todd M Foos
- Department of Molecular Biosciences, University of Kansas, Lawrence 66045, USA
| | | |
Collapse
|
30
|
Saransaari P, Oja SS. Modulation of taurine release by metabotropic receptors in the developing hippocampus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2002; 483:257-64. [PMID: 11787605 DOI: 10.1007/0-306-46838-7_28] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- P Saransaari
- Tampere Brain Research Center, University of Tampere Medical School, Finland
| | | |
Collapse
|
31
|
Wu JY, Chen W, Tang XW, Jin H, Foos T, Schloss JV, Davis K, Faiman MD, Hsu CC. Mode of action of taurine and regulation dynamics of its synthesis in the CNS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2002; 483:35-44. [PMID: 11787619 DOI: 10.1007/0-306-46838-7_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- J Y Wu
- Dept. of Mol Biosci., Univ. of Kansas, Lawrence 66045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Stover JF, Beyer TF, Unterberg AW. Riluzole reduces brain swelling and contusion volume in rats following controlled cortical impact injury. J Neurotrauma 2000; 17:1171-8. [PMID: 11186230 DOI: 10.1089/neu.2000.17.1171] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Modulation of the glutamatergic and excitotoxic pathway may attenuate secondary damage following traumatic brain injury by reducing presynaptic glutamate release and blocking sodium channels in their inactivated state. The aim of the present study was to investigate the neuroprotective potential of riluzole in traumatic brain-injured rats. A left temporoparietal contusion was induced in 70 male Sprague-Dawley rats (controlled cortical impact injury). Riluzole (8 mg/kg body weight) was given 30 min, and 6, 24, and 30 h after trauma, while control rats received physiological saline. Experiments were performed at two different degrees of trauma severity as defined by penetration depth of the impactor rod (1 vs. 1.5 mm) with the aim of investigating impact of severity of tissue damage on the neuroprotective potential of riluzole. At 48 h after trauma, brains were removed to determine hemispheric swelling and water content and to assess cortical contusion volume. Before brain removal cisternal cerebrospinal fluid (CSF) was collected in all rats to determine the effects of riluzole on substances associated with edema formation. For this, the excitatory transmitter glutamate, the volume-regulatory amino acid taurine, and the ATP-degradation product hypoxanthine were analyzed by high-performance liquid chromatography. Overall, the degree of tissue damage seems to influence the neuroprotective potential of riluzole. In rats with a less severe trauma (1-mm penetration depth), hemispheric swelling, cerebral water content of the traumatized hemisphere and cortical contusion volume were significantly reduced under riluzole compared to controls (p < 0.05). In rats with a more severe trauma (1.5-mm penetration depth), the neuroprotective effect of riluzole failed to reach statistical significance. Following trauma, CSF glutamate, taurine, and hypoxanthine levels were significantly increased compared to nontraumatized rats (p < 0.001). However, these neurochemical parameters as measured in cisternal CSF failed to reflect trauma-dependent increases in severity of tissue damage and did not reveal riluzole-mediated neuroprotection. Under the present study design, riluzole significantly reduced brain edema formation and contusion volume in rats subjected to a mild focal cortical contusion.
Collapse
Affiliation(s)
- J F Stover
- Charité-Department of Neurosurgery, Berlin, Germany.
| | | | | |
Collapse
|
33
|
Saransaari P, Oja SS. Modulation of the ischemia-induced taurine release by adenosine receptors in the developing and adult mouse hippocampus. Neuroscience 2000; 97:425-30. [PMID: 10828525 DOI: 10.1016/s0306-4522(00)00034-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The release of the inhibitory amino acid taurine is markedly enhanced under ischemic conditions in both adult and developing hippocampus, together with a pronounced increase in the release of excitatory amino acids and the neuromodulator adenosine. We studied the effects of adenosine receptor agonists and antagonists as well as adenosine transport inhibitors on hippocampal [(3)H]taurine release in normoxia and ischemia, using a superfusion system. Under standard conditions the adenosine A(1) receptor agonists N(6)-cyclohexyladenosine and R(-)N(6)-(2-phenylisopropyl)adenosine potentiated basal taurine release in developing mice and depressed the release in adults in a receptor-mediated manner. Adenosine A(2) receptor compounds had only minor effects on the basal release and the K(+)-stimulated release was not affected by these drugs. The adenosine uptake inhibitor dipyridamole enhanced basal taurine release in the developing hippocampus and reduced it in the adult. In ischemia the adenosine compounds had no marked effects on taurine release in immature animals, whereas A(1) receptor activation was still able to evoke taurine release in adults by a receptor-mediated mechanism. The results show that the basal release of taurine is modulated by A(1) receptors in both mature and immature hippocampus, whereas in ischemia these receptors potentiate taurine release only in adults. The elevated taurine levels together with the depression of excitatory amino acid release by adenosine receptor activation could be beneficial under ischemic conditions, protecting neural cells against excitotoxicity and hyperexcitation.
Collapse
Affiliation(s)
- P Saransaari
- Tampere Brain Research Center, University of Tampere Medical School, Box 607, FIN-33101, Tampere, Finland.
| | | |
Collapse
|
34
|
Stover JF, Unterberg AW. Increased cerebrospinal fluid glutamate and taurine concentrations are associated with traumatic brain edema formation in rats. Brain Res 2000; 875:51-5. [PMID: 10967298 DOI: 10.1016/s0006-8993(00)02597-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Glutamate-mediated excitotoxicity results in cell swelling and contributes to brain edema formation. Since increased extracellular taurine reflects glutamate-induced cell swelling in vitro, elevated CSF taurine could therefore unmask glutamate-mediated cytotoxic edema formation under in vivo conditions. For this, the temporal profile of brain edema and changes in cisternal CSF glutamate and taurine levels were determined in 28 rats following focal traumatic brain injury. Compared to six non-traumatized rats, CSF glutamate (4. 8+/-0.3 vs. 10+/-0.9 microM) and taurine levels (12+/-1.3 vs. 41+/-3 microM) were significantly increased at 8 h after trauma (P<0.001). Over time, CSF glutamate and taurine were significantly increased by 24 (glutamate: 38+/-4.4 microM) and 48 h (taurine: 51+/-4 microM), respectively. While CSF glutamate closely reflected changes in hemispheric water content, alterations in CSF taurine occurred diametrically to those seen for glutamate. Under the present study design, increased CSF taurine could reflect glutamate-induced cell swelling. In addition, neuronal release of taurine with its inhibitory and antiexcitotoxic functions could explain the observed diametric changes in CSF glutamate, CSF taurine, and hemispheric water content. Therefore, increasing taurine could be a therapeutic approach in attenuating post-traumatic glutamate-mediated cell damage.
Collapse
Affiliation(s)
- J F Stover
- Department of Neurosurgery, Charite-Virchow Medical Center, Augustenburger Platz 01, D-13353 Berlin, Germany.
| | | |
Collapse
|
35
|
del Olmo N, Bustamante J, del Río RM, Solís JM. Taurine activates GABA(A) but not GABA(B) receptors in rat hippocampal CA1 area. Brain Res 2000; 864:298-307. [PMID: 10802037 DOI: 10.1016/s0006-8993(00)02211-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We investigated if taurine, an endogenous GABA analog, could mimic both hyperpolarizing and depolarizing GABA(A)-mediated responses as well as pre- and postsynaptic GABA(B)-mediated actions in the CA1 region of rat hippocampal slices. Taurine (10 mM) perfusion induced changes in membrane potential and input resistance that are compatible with GABA(A) receptor activation. Local pressure application of taurine and GABA from a double barrel pipette positioned along the dendritic shaft of pyramidal cells revealed that taurine evoked a very small change of membrane potential and resistance compared with the large changes induced by GABA in these parameters. Moreover, in the presence of GABA(A) antagonists, local application of GABA on the dendrites evoked a GABA(B)-mediated hyperpolarization while taurine did not induce any change. Taurine neither mimicked baclofen inhibitory actions on presynaptic release of glutamate and GABA as judging by the lack of taurine effect on paired-pulse facilitation ratio and slow inhibitory postsynaptic potentials, respectively. These results show that taurine mainly activates GABA(A) receptors located on the cell body, indicating therefore that if taurine has any action on the dendrites it will not be mediated by either GABA(A) or GABA(B) receptors activation.
Collapse
Affiliation(s)
- N del Olmo
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, Ctra. de Colmenar Km 9, 28034, Madrid, Spain
| | | | | | | |
Collapse
|
36
|
del Olmo N, Galarreta M, Bustamante J, Martín del Rio R, Solís JM. Taurine-induced synaptic potentiation: role of calcium and interaction with LTP. Neuropharmacology 2000; 39:40-54. [PMID: 10665818 DOI: 10.1016/s0028-3908(99)00078-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Taurine induces a long-lasting potentiation of excitatory synaptic potentials due to the enhancement of both synaptic efficacy and axon excitability in the CA1 area of rat hippocampal slices. In this study, we characterized the role of Ca2+ in the generation of these long-lasting taurine effects. Taurine perfusion in a free-Ca2+ medium did not induce changes in either field excitatory synaptic potentials (fEPSP) slope or fiber volley (FV) amplitude. Intracellular recordings with a micropipette filled with the Ca2+ chelator BAPTA, prevented the EPSP potentiation induced by taurine in the impaled cell, whereas a long-lasting potentiation of the simultaneously recorded fEPSP was obtained. The depletion of intracellular Ca2+ stores by thapsigargin (1 microM), an inhibitor of endosomal Ca2+-ATPase, transformed the taurine-induced potentiation into a transitory process that declined to basal values after taurine withdrawal. Taurine-induced potentiation was not significantly affected by kynurenate (glutamate receptor antagonist), or nifedipine (high-voltage-activated Ca2+ channel antagonist). But, the presence of nickel (50 microM), an antagonist of low-voltage-activated Ca2+ channel, inhibited the taurine-induced potentiation, indicating that Ca2+ influx through this type of Ca2+ channels could account for the Ca2+ requirement of the taurine-induced potentiation. Occlusion experiments between tetanus-induced long-term potentiation (LTP) and taurine-induced potentiation indicate that both processes share some common mechanisms during the maintenance period.
Collapse
Affiliation(s)
- N del Olmo
- Departamento de Investigación, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | | | |
Collapse
|
37
|
Khan SH, Banigesh A, Baziani A, Todd KG, Miyashita H, Eweida M, Shuaib A. The role of taurine in neuronal protection following transient global forebrain ischemia. Neurochem Res 2000; 25:217-23. [PMID: 10786705 DOI: 10.1023/a:1007519419342] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Osmoregulation and post ischemic glutamate surge suppression (PIGSS) are important mechanisms in the neuroprotective properties of taurine. We studied the role of taurine in PIGSS following transient global forebrain ischemia (TGFI). A group of gerbils received a high dose of continuous intracerebral taurine during the peri-ischemic period. Beta-alanine was given similarly to a negative control group. The control group consisted of animals undergoing only TGFI. On the fourth day following commencement of drug administration, TGFI was induced. Concurrently, half the animals from each group receiving an agent had intracerebral microdialysis. All animals underwent histological assessment at day 7. The microdialysis and histological data was analyzed. Our results showed that taurine treatment did not cause PIGSS. The histological difference between the three groups was statistically insignificant. We conclude that intracerebral taurine in the dosage administered during peri-ischemic period, does not result in PIGSS or histologically evident neuroprotection.
Collapse
Affiliation(s)
- S H Khan
- Saskatchewan Stroke Research Centre, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | | | |
Collapse
|
38
|
Saransaari P, Oja SS. Characteristics of ischemia-induced taurine release in the developing mouse hippocampus. Neuroscience 1999; 94:949-54. [PMID: 10579587 DOI: 10.1016/s0306-4522(99)00384-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Taurine release in the developing hippocampus is markedly potentiated in ischemia. The mechanisms of the ischemia-induced release were studied in hippocampal slices from seven-day-old mice using a superfusion system. The basal release of [3H]taurine was significantly increased in media under normal conditions, but the ischemia-evoked release decreased in Na+ -free media, indicating the participation of Na+ -dependent transport processes. The involvement of taurine transporters in the release was confirmed with the structural analogs, hypotaurine and beta-alanine. These amino acids potentiated the release by trans-stimulation, but not in Na+ -free media. In the absence of Ca2+, the basal taurine release was markedly increased in normoxia but diminished in ischemia, indicating that a part of basal taurine release in ischemia is Ca2+ dependent. On the other hand, the K+ stimulation of taurine release was preserved in Ca2+ -free medium. The phospholipase and protein kinase inhibitors had no effect on ischemia-induced taurine release, nor did the chloride channel blockers 4-acetamido-4'-isothiocyanostilbene-2,2'-disulfonate (2 mM) and diisothiocyanostilbene-2,2'-disulfonate (0.1 mM) affect the release in ischemia. The increase in extracellular levels of taurine in the immature hippocampus in ischemia may serve as an important protective mechanism against excitotoxicity, to which the developing brain is particularly vulnerable, and contribute to the resistance of the immature brain to hypoxia.
Collapse
Affiliation(s)
- P Saransaari
- Tampere Brain Research Center, University of Tampere Medical School, Finland.
| | | |
Collapse
|
39
|
Saransaari P, Oja SS. Involvement of metabotropic glutamate receptors in taurine release in the adult and developing mouse hippocampus. Amino Acids 1999; 16:165-79. [PMID: 10319187 DOI: 10.1007/bf01321534] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The inhibitory amino acid taurine has been held to function as an osmoregulator and modulator of neural activity, being particularly important in the immature brain. Ionotropic glutamate receptor agonists are known markedly to potentiate taurine release. The effects of different metabotropic glutamate receptor (mGluR) agonists and antagonists on the basal and K(+)-stimulated release of [3H]taurine from hippocampal slices from 3-month-old (adult) and 7-day-old mice were now investigated using a superfusion system. Of group I metabotropic glutamate receptor agonists, quisqualate potentiated basal taurine release in both age groups, more markedly in the immature hippocampus. This action was not antagonized by the specific antagonists of group I but by 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and 6-nitro-7-sulphamoylbenzo[f]quinoxaline-2,3-dione (NBQX), which would suggest an involvement of ionotropic glutamate receptors. (S)-3,5-dihydroxyphenylglycine (DHPG) potentiated the basal release by a receptor-mediated mechanism in the immature hippocampus. The group II agonist (2S, 2'R, 3'R)-2-(2',3'-dicarboxycyclopropyl)glycine (DCG IV) markedly potentiated basal taurine release at both ages. These effects were antagonized by dizocilpine, indicating again the participation of ionotropic receptors. Group III agonists slightly potentiated basal taurine release, as did several antagonists of the three metabotropic receptor groups. Potassium-stimulated (50 mM K+) taurine release was generally significantly reduced by mGluR agents, mainly by group I and II compounds. This may be harmful to neurons in hyperexcitatory states. On the other hand, the potentiation by mGluRs of basal taurine release, particularly in the immature hippocampus, together with the earlier demonstrated pronounced enhancement by activation of ionotropic glutamate receptors, may protect neurons against excitotoxicity.
Collapse
Affiliation(s)
- P Saransaari
- Tampere Brain Research Center, University of Tampere Medical School, Finland.
| | | |
Collapse
|
40
|
Stover JF, Morganti-Kosmann MC, Lenzlinger PM, Stocker R, Kempski OS, Kossmann T. Glutamate and taurine are increased in ventricular cerebrospinal fluid of severely brain-injured patients. J Neurotrauma 1999; 16:135-42. [PMID: 10098958 DOI: 10.1089/neu.1999.16.135] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Glutamate contributes to secondary brain damage, resulting in cell swelling and brain edema. Under in vitro conditions, increased extracellular levels of the amino acid taurine reflect glutamate-induced osmotic cell swelling. In vivo, increases in cerebrospinal fluid (CSF) taurine could, therefore, unmask glutamate-mediated cytotoxic edema formation and possibly differentiate it from vasogenic edema. To test this hypothesis, ventricular CSF glutamate and taurine levels were measured in 28 severely brain-injured patients on days 1, 5, and 14 after trauma. Posttraumatic changes in CSF amino acids were investigated in regard to extent of tissue damage and alterations in brain edema as estimated by computerized tomography. On day 1, CSF glutamate and taurine levels were significantly increased in patients with subdural or epidural hematomas (8+/-0.8/71+/-12 microM), contusions (21+/-4.1/122+/-18 microM), and generalized brain edema (13+/-3.2/80+/-15 microM) compared to lumbar control CSF (1.3+/-0.1/12+/-1 microM; p < 0.001). CSF amino acids, however, did not reflect edema formation and resolution as estimated by computerized tomography. CSF taurine correlated positively with glutamate, eventually depicting glutamate-induced cell swelling. However, parallel neuronal release of taurine with its inhibitory function cannot be excluded. Thus, the sensitivity of taurine in unmasking cytotoxic edema formation is weakened by the inability in defining its origin and function under the conditions chosen in the present study. Overall, persisting pathologic ventricular CSF glutamate and taurine levels are highly suggestive of ongoing glial and neuronal impairment in humans following severe traumatic brain injury.
Collapse
Affiliation(s)
- J F Stover
- Department of Trauma Surgery, University Hospital Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
41
|
Hada J, Kaku T, Morimoto K, Hayashi Y, Nagai K. Activation of adenosine A2 receptors enhances high K(+)-evoked taurine release from rat hippocampus: a microdialysis study. Amino Acids 1999; 15:43-52. [PMID: 9871486 DOI: 10.1007/bf01345279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The present study was designed to examine which type of adenosine receptors was involved in enhancement of high K(+)-evoked taurine release from in vivo rat hippocampus using microdialysis. Perfusion with 0.5 or 5.0 mM adenosine enhanced high K(+)-evoked taurine release. Perfusion with 2 microM R(-)-N6-2-phenylisopropyladenosine (PIA), a selective adenosine A1 receptor agonist, did not modulate taurine release. Perfusion with 1 microM 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), a selective adenosine A1 receptor antagonist, increased taurine release. On the other hand, perfusion with 20 microM 2-[4-(2-carboxyethyl)phenethylamino]-5'-N-ethyl-carboxamide-adenos ine (CGS21680), a selective adenosine A2A receptor agonist, enhanced taurine release, while perfusion with 1 mM 3,7-dimethyl-propagylxanthine (DMPX), an adenosine A2 receptor antagonist, did not affect taurine release. These results demonstrate that adenosine enhances high K(+)-evoked taurine release via activation of adenosine A2A receptors from both neurons and glial cells of in vivo rat hippocampus.
Collapse
Affiliation(s)
- J Hada
- Department of Physiology, Hyogo College of Medicine, Japan
| | | | | | | | | |
Collapse
|
42
|
Abstract
Estrogen deficiency, hyperinsulinemia, type II diabetes, atherosclerosis, and a past history of elevated blood pressure may be associated with increased risk of Alzheimer's disease (AD). Common to all of these risk factors is a diminished capacity of vascular endothelium to generate nitric oxide (NO). Vascular NO has the potential to enhance the membrane polarization of cerebral neurons by increasing the open probability of calcium-activated potassium channels; this may protect neurons from the excessive calcium influx, potentiated by beta-amyloid peptides that is thought to mediate neuronal damage in AD. The possibility that NO/cyclic guanosine 3', 5'-phosphate (cGMP) may modulate the synthesis or processing of the amyloid precursor protein, also merits evaluation. Practical measures for promoting vascular NO production may include increased intakes of arginine, potassium, antioxidants, and fish-oil, as well as lifestyle measures that typically lower elevated blood pressure; potential benefits of chromium, glucosamine, and silicon should also be explored. In hypertensives, angiotensin-converting enzyme (ACE) inhibitors and sodium restriction may favorably influence endothelial function. Fish-oil should have the additional benefit of antagonizing the contribution of interleukin-1 to AD pathogenesis. Ancillary anti-excitotoxic measures such as magnesium, taurine, phenytoin, and vasodilators targeting ATP-dependent potassium (KATP) channels, may likewise reduce AD risk. Most of the nutritional measures suggested here would in any case be recommendable for preservation of vascular health.
Collapse
|
43
|
Wu JY, Tang XW, Schloss JV, Faiman MD. Regulation of taurine biosynthesis and its physiological significance in the brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998; 442:339-45. [PMID: 9635049 DOI: 10.1007/978-1-4899-0117-0_42] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cysteine sulfinic acid decarboxylase (CSAD), the rate-limiting enzyme in taurine biosynthesis, was found to be activated under conditions that favor protein phosphorylation and inactivated under conditions favoring protein dephosphorylation. Direct incorporation of 32P into purified CSAD has been demonstrated with [gamma 32P]ATP and PKC, but not PKA. In addition, the 32P labeling of CSAD was inhibited by PKC inhibitors suggesting that PKC is responsible for phosphorylation of CSAD in the brain. Okadaic acid had no effect on CSAD activity at 10 microM suggesting that protein phosphatase-2C (PrP-2C) might be involved in the dephosphorylation of CSAD. Furthermore, it was found that either glutamate- or high K(+)-induced depolarization increased CSAD activity as well as 32P-incorporation into CSAD in neuronal cultures, supporting the notion that the CSAD activity is endogenously regulated by protein phosphorylation in the brain. A model to link neuronal excitation, phosphorylation of CSAD and increase in taurine biosynthesis is proposed.
Collapse
Affiliation(s)
- J Y Wu
- Department of Physiology and Cell Biology, University of Kansas, Lawrence 66045, USA
| | | | | | | |
Collapse
|
44
|
Satoh H, Sperelakis N. Review of some actions of taurine on ion channels of cardiac muscle cells and others. GENERAL PHARMACOLOGY 1998; 30:451-63. [PMID: 9522160 DOI: 10.1016/s0306-3623(97)00309-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Taurine has recently been known to protect against ischemia and heart failure. Taurine possesses plenty of actions on the ion channels and transports, but is very non-specific. 2. Taurine may directly and indirectly help to regulate the [Ca]i level by modulating the activity of the voltage-dependent Ca2+ channels (also dependent on [Ca]i/[Ca]o), by regulation of Na+ channels, and secondly via Na-Ca exchange and Na(+)-taurine cotransport. 3. Taurine can prevent the Ca2+ ([Ca]o or [Ca]i)-induced cardiac functions. 4. Therefore, it seems possible that taurine could exert the potent cardioprotective actions even under the condition of low [Ca]i levels as well as under the Ca2+ overload condition. 5. The electrophysiological actions of taurine on cardiomyocytes, smooth muscle cells, and neurons from recent studies are summarized.
Collapse
Affiliation(s)
- H Satoh
- Department of Pharmacology, Nara Medical University, Japan
| | | |
Collapse
|
45
|
Abstract
Taurine is known to be involved in many important physiological functions. Here we report that both in vivo and in vitro the taurine-synthesizing enzyme in the brain, namely cysteine sulfinic acid decarboxylase (CSAD), is activated when phosphorylated and inhibited when dephosphorylated. Furthermore, protein kinase C and protein phosphatase 2C have been identified as the enzymes responsible for phosphorylation and dephosphorylation of CSAD, respectively. In addition, the effect of neuronal depolarization on CSAD activity and 32P incorporation into CSAD in neuronal cultures is also included. A model to link neuronal excitation and CSAD activation by a Ca2+-dependent protein kinase is proposed.
Collapse
|
46
|
Glutamate-agonist-evoked taurine release from the adult and developing mouse hippocampus in cell-damaging conditions. Amino Acids 1997. [DOI: 10.1007/bf01372596] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Miyata S, Matsushima O, Hatton GI. Taurine in rat posterior pituitary: Localization in astrocytes and selective release by hypoosmotic stimulation. J Comp Neurol 1997. [DOI: 10.1002/(sici)1096-9861(19970519)381:4<513::aid-cne10>3.0.co;2-j] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
48
|
Lubec B, Ya-hua Z, Pertti S, Pentti T, Kitzmüller E, Lubec G. Distribution and disappearance of the radiolabeled carbon derived from L-arginine and taurine in the mouse. Life Sci 1997; 60:2373-81. [PMID: 9199481 DOI: 10.1016/s0024-3205(97)00297-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
L-arginine and taurine are still in the center of physiological and pharmacological research. Although the fate of nitrogen of both compounds and of the 35S-taurine is well-documented, the fate of the carbon skeleton has not been elucidated yet. We studied the organ distribution of 14C arginine and 14C taurine over time in the mouse using whole body autoradiography with densitometric image analysis. We describe different organ distribution patterns. Kidney, heart, lung, the Harderian gland, the central nervous system, intestine and testis showed a comparable pattern of arginine disappearance in contrast to rapid disappearance in the salivary gland and the accumulation pattern in bone and spleen. Data on 14C taurine of liver, kidneys, lung, testis and Harderian gland resembled the arginine pattern; Accumulation of taurine carbon was found in salivary gland, bone, intestine, heart and brain. Our studies challenge and demand further related studies to obtaining more information on the fate of the carbon skeleton of these amino acids.
Collapse
Affiliation(s)
- B Lubec
- University of Vienna, Dpt of Neonatology, Austria
| | | | | | | | | | | |
Collapse
|
49
|
Tang X, Hsu CC, Sun Y, Wu E, Yang CY, Wu JY. Multiplicity of Brain Cysteine Sulfinic Acid Decarboxylase - Purification, Characterization and Subunit Structure. J Biomed Sci 1996; 3:442-453. [PMID: 11725125 DOI: 10.1007/bf02258048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Cysteine sulfinic acid decarboxylase (CSAD), the rate-limiting enzyme in taurine biosynthesis, appears to be present in the brain in multiple isoforms. Two distinct forms of CSAD, referred to as CSAD I and CSAD II, were obtained on Sephadex G-100 column. CSAD I and CSAD II differ in: (1) the elution profile on Sephadex G-100 column; (2) the sensitivity towards Mn(2+), methione, and other sulfur-containing amino acids, and (3) their immunologic properties. CSAD II has been purified to about 2,500-fold by a combination of column chromatographies and polyacrylamide gel electrophoresis (PAGE). The purity of the enzyme preparation was established as judged from the following observations: (1) a single protein band was observed under various electrophoretic conditions, e.g., 5-20% nondenaturing PAGE, 7% nondenaturing PAGE and 10% SDS-PAGE, and (2) in nondenaturing PAGE, the protein band comigrated with CSAD activity. CSAD II has a molecular weight of 90 kDa and is a homodimer consisting of two 43 +/- 2 kDa subunits. CSAD appears to require Mn(2+) for its maximum activity. Other divalent cations fail to replace Mn(2+) in activation of CSAD activity. However, the precise role of Mn(2+) in the action of CSAD remains to be determined. Copyright 1996 S. Karger AG, Basel
Collapse
Affiliation(s)
- X.W. Tang
- Department of Physiology and Cell Biology, University of Kansas, Lawrence, Kans., USA
| | | | | | | | | | | |
Collapse
|
50
|
Hada J, Kaku T, Morimoto K, Hayashi Y, Nagai K. Adenosine transport inhibitors enhance high K(+)-evoked taurine release from rat hippocampus. Eur J Pharmacol 1996; 305:101-7. [PMID: 8813538 DOI: 10.1016/0014-2999(96)00171-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We examined the effects of Ca(2+)-free medium containing 20 mM Mg2+, a non-selective adenosine receptor antagonist, theophylline, and adenosine transport inhibitors, dipyridamole and nitrobenzylthioinosine, on high K(+)-evoked spreading depression, glutamate, and taurine release from the rat hippocampus using brain microdialysis. High K+ alone perfusion evoked spreading depression and increased glutamate release followed by taurine efflux. Perfusion of Ca(2+)-free medium with high K+ never evoked spreading depression and decreased the high K(+)-evoked taurine release. Perfusion of theophylline (1 mM) increased the occurrence of high K(+)-evoked spreading depression and glutamate release, but did not modify taurine release. In contrast, simultaneous perfusion of dipyridamole (100 microM) and nitrobenzylthioinosine (50 microM) reduced the occurrence of spreading depression and the high K(+)-evoked glutamate release, but enhanced significantly the taurine efflux. These findings suggest that endogenous taurine with adenosine may have neuroprotective actions against high K(+)-evoked glutamate release and spreading depression in the rat hippocampus, in addition to its osmoregulatory action.
Collapse
Affiliation(s)
- J Hada
- Department of Physiology, Hyogo College of Medicine, Japan
| | | | | | | | | |
Collapse
|