1
|
Yuwattana R, Suparan K, Kerdphoo S, Arunsak B, Sanguansermsri C, Katanyuwong K, Chattipakorn N, Wiwattanadittakul N, Chattipakorn SC. Altered gut microbiome profiles in epileptic children are associated with spectrum of anti-seizure medication responsiveness. Brain Res 2025; 1849:149367. [PMID: 39626831 DOI: 10.1016/j.brainres.2024.149367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024]
Abstract
Gut microbiota plays a role in epilepsy. However, current knowledge of how gut dysbiosis is associated with a response to anti-seizure medications (ASMs) in epileptic children is still limited. We aimed to characterize the gut microbiota profiles in epileptic children based on response to ASMs. Eighty-six children aged 3-18 years old with a regular oral diet were enrolled onto the study and divided into three groups in accordance with ILAE definitions: 26 healthy controls, 31 drug-sensitive epilepsy (DSE) patients, and 29 drug-resistant epilepsy (DRE) patients. Based on ASM responsiveness, defined as a reduction in seizure frequency of at least 75 % over one year, DRE individuals were subclassified into 13 drug responsive (DRE-DR) and 16 drug non-responsive (DRE-DNR) patients. Feces were collected at the time of enrollment for gut microbiota analysis using 16S rRNA sequencing. Epileptic patients exhibited distinctive gut dysbiotic profiles. Differential abundance investigation revealed that CAG-56 was significantly increased in epileptic patients compared to controls. Saccharimonadales and Peptoclostridium significantly increased in the DSE group, compared to the DRE group. Vibrionaceae, especially Grimontia, Rhodobacteraceae, and Enterobacter were significantly abundant in the DRE-DNR group, followed by abundance in the DRE-DR and DSE groups. Outcomes from PICRUSt2 analysis predicted that epileptic patients, especially those in the DRE group, had increased metabolic pathways responsible for vanillin and taurine degradation, compared to controls. These findings suggest that gut dysbiosis could play roles in epileptogenesis and ASM resistance. Notably, the identified gut microbes could serve as predictive biomarkers for the DRE condition.
Collapse
Affiliation(s)
- Rattakarn Yuwattana
- Division of Neurology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokphong Suparan
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chinnuwat Sanguansermsri
- Division of Neurology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kamornwan Katanyuwong
- Division of Neurology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Natrujee Wiwattanadittakul
- Division of Neurology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
2
|
Ali I, Jupp B, Hudson MR, Major B, Silva J, Yamakawa GR, Casillas-Espinosa PM, Braine E, Thergarajan P, Haskali MB, Vivash L, Brkljaca R, Shultz SR, Kwan P, Fukushima K, Sachdev P, Cheng JY, Mychasiuk R, Jones NC, Wright DK, OBrien TJ. In vivo biomarkers of GABAergic function in epileptic rats treated with the GAT-1 inhibitor E2730. Epilepsia 2024; 65:3376-3390. [PMID: 39302665 DOI: 10.1111/epi.18119] [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: 05/21/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE E2730, an uncompetitive γ-aminobutyric acid (GABA) transporter-1 (GAT-1) inhibitor, has potent anti-seizure effects in a rodent model of chronic temporal lobe epilepsy, the kainic acid status epilepticus (KASE) rat model. In this study, we examined purported neuroimaging and physiological surrogate biomarkers of the effect of E2730 on brain GABAergic function. METHODS We conducted a randomized cross-over study, incorporating 1-week treatments with E2730 (100 mg/kg/day subcutaneous infusion) or vehicle in epileptic post-KASE rats. KASE rats underwent serial 9.4 T magnetic resonance spectroscopy (MRS) measuring GABA and other brain metabolites, [18F]Flumazenil positron emission tomography (PET) quantifying GABAA receptor availability, quantitative electroencephalography (qEEG) and transcranial magnetic stimulation (TMS)-mediated motor activity, as well as continuous video-EEG recording to measure spontaneous seizures during each treatment. Age-matched, healthy control animals treated with E2730 or vehicle were also studied. RESULTS E2730 treatment significantly reduced spontaneous seizures, with 8 of 11 animals becoming seizure-free. MRS revealed that E2730-treated animals had significantly reduced taurine levels. [18F]Flumazenil PET imaging revealed no changes in GABA receptor affinity or density during E2730 treatment. The power of gamma frequency oscillations in the EEG was decreased significantly in the auditory cortex and hippocampus of KASE and control rats during E2730 treatment. Auditory evoked gamma frequency power was enhanced by E2730 treatment in the auditory cortex of KASE and healthy controls, but only in the hippocampus of KASE rats. E2730 did not influence motor evoked potentials triggered by TMS. SIGNIFICANCE This study identified clinically relevant changes in multimodality imaging and functional purported biomarkers of GABAergic activity during E2730 treatment in epileptic and healthy control animals. These biomarkers could be utilized in clinical trials of E2730 and potentially other GABAergic drugs to provide surrogate endpoints, thereby reducing the cost of such trials.
Collapse
Affiliation(s)
- Idrish Ali
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Bianca Jupp
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Matthew R Hudson
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Brendan Major
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Juliana Silva
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn R Yamakawa
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Pablo M Casillas-Espinosa
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Emma Braine
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | | | - Mohammad B Haskali
- Department of Radiopharmaceutical Sciences, Cancer Imaging, The Peter MacCallum Cancer Centre, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Lucy Vivash
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | | | - Sandy R Shultz
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, Canada
| | - Patrick Kwan
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | | | - Pallavi Sachdev
- Clinical Evidence Generation, Translational Sciences, Eisai Inc., Bunkyo, Japan
| | - Jocelyn Y Cheng
- Clinical Evidence Generation, Translational Sciences, Eisai Inc., Bunkyo, Japan
| | | | - Nigel C Jones
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - David K Wright
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Terence J OBrien
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Akiyama T, Saigusa D, Inoue T, Tokorodani C, Akiyama M, Michiue R, Mori A, Hishinuma E, Matsukawa N, Shibata T, Tsuchiya H, Kobayashi K. Exploration of urine metabolic biomarkers for new-onset, untreated pediatric epilepsy: A gas and liquid chromatography mass spectrometry-based metabolomics study. Brain Dev 2024; 46:180-186. [PMID: 38171994 DOI: 10.1016/j.braindev.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/02/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE The discovery of objective indicators for recent epileptic seizures will help confirm the diagnosis of epilepsy and evaluate therapeutic effects. Past studies had shortcomings such as the inclusion of patients under treatment and those with various etiologies that could confound the analysis results significantly. We aimed to minimize such confounding effects and to explore the small molecule biomarkers associated with the recent occurrence of epileptic seizures using urine metabolomics. METHODS This is a multicenter prospective study. Subjects included pediatric patients aged 2 to 12 years old with new-onset, untreated epilepsy, who had had the last seizure within 1 month before urine collection. Controls included healthy children aged 2 to 12 years old. Those with underlying or chronic diseases, acute illnesses, or recent administration of medications or supplements were excluded. Targeted metabolome analysis of spot urine samples was conducted using gas chromatography (GC)- and liquid chromatography (LC)-tandem mass spectrometry (MS/MS). RESULTS We enrolled 17 patients and 21 controls. Among 172 metabolites measured by GC/MS/MS and 41 metabolites measured by LC/MS/MS, only taurine was consistently reduced in the epilepsy group. This finding was subsequently confirmed by the absolute quantification of amino acids. No other metabolites were consistently altered between the two groups. CONCLUSIONS Urine metabolome analysis, which covers a larger number of metabolites than conventional biochemistry analyses, found no consistently altered small molecule metabolites except for reduced taurine in epilepsy patients compared to healthy controls. Further studies with larger samples, subjects with different ages, expanded target metabolites, and the investigation of plasma samples are required.
Collapse
Affiliation(s)
- Tomoyuki Akiyama
- Department of Pediatrics (Child Neurology), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takushi Inoue
- Department of Pediatric Neurology, NHO Okayama Medical Center, Okayama, Japan
| | - Chiho Tokorodani
- Department of Pediatrics, Kochi Health Sciences Center, Kochi, Japan
| | - Mari Akiyama
- Department of Pediatrics (Child Neurology), Okayama University Hospital, Okayama, Japan
| | - Rie Michiue
- Department of Pediatrics (Child Neurology), Okayama University Hospital, Okayama, Japan
| | - Atsushi Mori
- Department of Neurology, Shiga Medical Center for Children, Moriyama, Japan
| | - Eiji Hishinuma
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Advanced Research Centre for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Naomi Matsukawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takashi Shibata
- Department of Pediatrics (Child Neurology), Okayama University Hospital, Okayama, Japan
| | - Hiroki Tsuchiya
- Department of Pediatrics (Child Neurology), Okayama University Hospital, Okayama, Japan
| | - Katsuhiro Kobayashi
- Department of Pediatrics (Child Neurology), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
4
|
Meera P, Uusi-Oukari M, Lipshutz GS, Wallner M. GABA A receptors as plausible molecular targets and mediators for taurine and homotaurine actions. Front Pharmacol 2023; 14:1271203. [PMID: 38155909 PMCID: PMC10752957 DOI: 10.3389/fphar.2023.1271203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Dementia and autoimmune diseases are prevalent conditions with limited treatment options. Taurine and homotaurine (HT) are naturally occurring sulfonate amino acids, with taurine being highly abundant in animal tissues, but declining with age in the blood. HT is a blood-brain barrier permeable drug under investigation for Alzheimer's disease. HT also has beneficial effects in a mouse model of multiple sclerosis likely through an anti-inflammatory mechanism mediated by GABAA receptor (GABAAR) agonism in immune cells. While both taurine and HT are structural GABA analogs and thought to be GABA mimetics at GABAARs, there is uncertainty concerning their potency as GABA mimetics on native GABAARs. We show that HT is a very potent GABA mimetic, as it evokes GABAAR-mediated currents with an EC50 of 0.4 μM (vs. 3.7 μM for GABA and 116 µM for taurine) in murine cerebellar granule cells in brain slices, with both taurine and HT having similar efficacy in activating native GABAARs. Furthermore, HT displaces the high affinity GABAAR ligand [3H]muscimol at similarly low concentrations (HT IC50 of 0.16 μM vs. 125 μM for taurine) in mouse brain homogenates. The potency of taurine and HT as GABAAR agonists aligns with endogenous concentrations of taurine in the blood and with HT concentrations achieved in the brain following oral administration of HT or the HT pro-drug ALZ-801. Consequently, we discuss that GABAARs subtypes, similar to the ones we studied here in neurons, are plausible targets for mediating the potential beneficial effects of taurine in health and life-span extension and the beneficial HT effects in dementia and autoimmune conditions.
Collapse
Affiliation(s)
- Pratap Meera
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mikko Uusi-Oukari
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Gerald S. Lipshutz
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
- Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA, United States
| | - Martin Wallner
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
5
|
Kaur A, Kumar S, Goel RK. Adjunct antiseizure effect of clotrimazole in a rotenone corneal kindling mouse model of mitochondrial drug-resistant epilepsy. Epilepsy Res 2023; 198:107246. [PMID: 37925976 DOI: 10.1016/j.eplepsyres.2023.107246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
This study aimed to investigate the therapeutic potential of clotrimazole, an inhibitor of the transient receptor potential cation channel, for treating mitochondrial drug-resistant epilepsy and to understand its underlying neurochemical mechanisms. Adult albino mice underwent rotenone-corneal kindling, receiving daily electric shocks (15 mA, 20 V, 6-Hz for 3 s) through a corneal electrode, to induce mitochondrial drug-resistant epilepsy. The onset of drug resistance was confirmed by the significant (p < 0.05) lack of seizure control with standard antiseizure medications including levetiracetam (40 mg/kg), valproate (250 mg/kg), phenytoin (35 mg/kg), lamotrigine (15 mg/kg), and carbamazepine (40 mg/kg). Drug-resistant mice were then classified into one vehicle-treated group and three groups treated with varying doses of clotrimazole (40, 80, and 160 mg/kg orally). Neurochemical analysis of the seizurogenic hippocampus and cerebral cortex was conducted using high-performance liquid chromatography with an electrochemical detector. Administration of clotrimazole alongside standard antiseizure medications led to a significant decrease (p < 0.05) in seizure scores suggesting the restoration of antiseizure effects. Neurochemicals, including tryptophan, serotonin, kynurenine, serine, taurine, gamma-aminobutyric acid, and glutamate, were significantly restored post-clotrimazole treatment. Overall, the present study underscores the adjunct antiseizure effect of clotrimazole in a rotenone corneal kindling mouse model of mitochondrial drug-resistant epilepsy, emphasising its role in neurochemical restoration.
Collapse
Affiliation(s)
- Arvinder Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Sandeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India; Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Rajesh Kumar Goel
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, Punjab, India.
| |
Collapse
|
6
|
Santulli G, Kansakar U, Varzideh F, Mone P, Jankauskas SS, Lombardi A. Functional Role of Taurine in Aging and Cardiovascular Health: An Updated Overview. Nutrients 2023; 15:4236. [PMID: 37836520 PMCID: PMC10574552 DOI: 10.3390/nu15194236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Taurine, a naturally occurring sulfur-containing amino acid, has attracted significant attention in recent years due to its potential health benefits. Found in various foods and often used in energy drinks and supplements, taurine has been studied extensively to understand its impact on human physiology. Determining its exact functional roles represents a complex and multifaceted topic. We provide an overview of the scientific literature and present an analysis of the effects of taurine on various aspects of human health, focusing on aging and cardiovascular pathophysiology, but also including athletic performance, metabolic regulation, and neurological function. Additionally, our report summarizes the current recommendations for taurine intake and addresses potential safety concerns. Evidence from both human and animal studies indicates that taurine may have beneficial cardiovascular effects, including blood pressure regulation, improved cardiac fitness, and enhanced vascular health. Its mechanisms of action and antioxidant properties make it also an intriguing candidate for potential anti-aging strategies.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Fahimeh Varzideh
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Pasquale Mone
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Stanislovas S. Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| |
Collapse
|
7
|
Pawlik M, Czarnecka AM, Kołodziej M, Skowrońska K, Węgrzynowicz M, Podgajna M, Czuczwar SJ, Albrecht J. Attenuation of initial pilocarpine-induced electrographic seizures by methionine sulfoximine pretreatment tightly correlates with the reduction of extracellular taurine in the hippocampus. Epilepsia 2023; 64:1390-1402. [PMID: 36808593 DOI: 10.1111/epi.17554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/20/2023]
Abstract
OBJECTIVE Initiation and development of early seizures by chemical stimuli is associated with brain cell swelling resulting in edema of seizure-vulnerable brain regions. We previously reported that pretreatment with a nonconvulsive dose of glutamine (Gln) synthetase inhibitor methionine sulfoximine (MSO) mitigates the intensity of initial pilocarpine (Pilo)-induced seizures in juvenile rats. We hypothesized that MSO exerts its protective effect by preventing the seizure-initiating and seizure-propagating increase of cell volume. Taurine (Tau) is an osmosensitive amino acid, whose release reflects increased cell volume. Therefore, we tested whether the poststimulus rise of amplitude of Pilo-induced electrographic seizures and their attenuation by MSO are correlated with the release of Tau from seizure-affected hippocampus. METHODS Lithium-pretreated animals were administered MSO (75 mg/kg ip) 2.5 h before the induction of convulsions by Pilo (40 mg/kg ip). Electroencephalographic (EEG) power was analyzed during 60 min post-Pilo, at 5-min intervals. Extracellular accumulation of Tau (eTau) served as a marker of cell swelling. eTau, extracellular Gln (eGln), and extracellular glutamate (eGlu) were assayed in the microdialysates of the ventral hippocampal CA1 region collected at 15-min intervals during the whole 3.5-h observation period. RESULTS The first EEG signal became apparent at ~10 min post-Pilo. The EEG amplitude across most frequency bands peaked at ~40 min post-Pilo, and showed strong (r ~ .72-.96) temporal correlation with eTau, but no correlation with eGln or eGlu. MSO pretreatment delayed the first EEG signal in Pilo-treated rats by ~10 min, and depressed the EEG amplitude across most frequency bands, to values that remained strongly correlated with eTau (r > .92) and moderately correlated (r ~ -.59) with eGln, but not with eGlu. SIGNIFICANCE Strong correlation between attenuation of Pilo-induced seizures and Tau release indicates that the beneficial effect of MSO is due to the prevention of cell volume increase concurrent with the onset of seizures.
Collapse
Affiliation(s)
- Marek Pawlik
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maria Czarnecka
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marcin Kołodziej
- Institute of Theory of Electrical Engineering, Measurement, and Information Systems, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Michał Węgrzynowicz
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Martyna Podgajna
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
8
|
Hosoi Y, Akita T, Watanabe M, Ito T, Miyajima H, Fukuda A. Taurine depletion during fetal and postnatal development blunts firing responses of neocortical layer II/III pyramidal neurons. Front Mol Neurosci 2022; 15:806798. [PMID: 36466806 PMCID: PMC9712787 DOI: 10.3389/fnmol.2022.806798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 10/27/2022] [Indexed: 08/22/2023] Open
Abstract
Fetal and infant brains are rich in maternally derived taurine. We previously demonstrated that taurine action regulates the cation-chloride cotransporter activity and the differentiation and radial migration of pyramidal neuron progenitors in the developing neocortex of rodent fetuses. Here we examined the effects of fetal and infantile taurine depletion caused by knockout of the taurine transporter Slc6a6 on firing properties of layer II/III pyramidal neurons in the mouse somatosensory cortex at 3 weeks of postnatal age, using the whole-cell patch-clamp technique. The membrane excitability under resting conditions was similar between the neurons in knockout mice and those in wildtype littermates. However, the frequency of repetitive spike firing during moderate current injection was significantly lower, along with lower membrane voltage levels during interspike intervals in knockout neurons. When strong currents were injected, by which repetitive firing was rapidly abolished due to inactivation of voltage-gated Na+ channels in wildtype neurons, the firing in knockout neurons lasted for a much longer period than in wildtype neurons. This was due to much lower membrane voltage levels during interspike intervals in knockout neurons, promoting greater recovery of voltage-gated Na+ channels from inactivation. Thus, taurine depletion in pyramidal neurons blunted neuronal responses to external stimuli through increasing the stability of repetitive firing, presumably mediated by larger increases in membrane K+ conductance during interspike intervals.
Collapse
Affiliation(s)
- Yasushi Hosoi
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tenpei Akita
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Division of Health Science, Department of Basic Nursing, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takashi Ito
- Department of Biosciences and Biotechnology, Fukui Prefectural University, Fukui, Japan
| | - Hiroaki Miyajima
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
9
|
Bridging potential of Taurine-loading PCL conduits transplanted with hEnSCs on resected sciatic nerves. Regen Ther 2022; 21:424-435. [PMID: 36274680 PMCID: PMC9556906 DOI: 10.1016/j.reth.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 11/05/2022] Open
Abstract
Reconstruction of nerve conduits is a promising method for functional improvement in peripheral nerve repair. Besides choosing of a suitable polymer for conduit construction, adding factors such as Taurine improve a more advantageous microenvironment for defect nerve regeneration. Showing several major biological properties of Taurine, for example, regulation of the osmotic pressure, modulation of neurogenesis, and calcium hemostasis, makes it an appropriate option for repairing of defected nerves. To this, we examined repairing effects of Taurine-loading PCL conduits cultured with human endothelial stem cells (hEnSCs) on resected sciatic nerves. PCL/Taurine/Cell conduits transplanted to a 10-mm sciatic nerve gap. Forty-two wistar rats were randomly divided to seven groups: (1) Normal group, (2) Negative control (NC), (3) Positive control (nerve Autograft group), (4) PCL conduits group (PCL), (5) Taurine loaded PCL conduits group (PCL/Taurine), (6) hEnSCs cultured on the PCL conduits (PCL/Cell), (7) hEnSCs cultured on the PCL/Taurine conduits (PCL/Taurine/Cell). Functional recovery of motor and sensory nerves, the action potential of exciting muscle and motor distal latency has seen in PCL/Taurine/Cell conduits. Histological studies showed also remarkable nerve regeneration and obvious bridging has seen in this group. In conclusion, PCL/Taurine/Cell conduits showing suitable mechanical properties and biocompatibility may improve sciatic nerve regeneration.
Collapse
Key Words
- AD, Alzheimer's disease
- DAPI, diamidino phenylindole
- DPN, peripheral neuropathy
- ECM, extracellular matrix structure
- EMAP, muscle action potential
- EMG, electromyography
- FBS, fetal bovine serum
- FDA, Food and Drug Administration
- HPF, high power fields
- HPL, hotplate latency
- Human endothelial stem cells (hEnSCs)
- LFB, Luxol fast blue
- MSCs, mesenchymal stem cells
- MTT, dimethylthiazol diphenyl tetrazolium bromide
- NGC, nerve guidance conduits
- Nerve regeneration
- PBS, phosphate-buffered saline
- PCL, polycaprolactone
- PD, Parkinson's disease
- PNS, peripheral nerve system
- SFI, sciatic functionl index
- TCP, tissue culture plate
- Taurine
- WRL, withdrawal reflex latency
- hEnSCs, human endothelial stem cells
Collapse
|
10
|
Lee M, Kim EJ, Kim MJ, Yum MS. Rapamycin Cannot Reduce Seizure Susceptibility in Infantile Rats with Malformations of Cortical Development Lacking mTORC1 Activation. Mol Neurobiol 2022; 59:7439-7449. [PMID: 36194361 DOI: 10.1007/s12035-022-03033-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022]
Abstract
The mechanistic target of the rapamycin (mTOR) pathway is involved in cortical development. However, the efficacy of mTOR inhibitors in malformations of cortical dysplasia (MCD) outside of the tuberous sclerosis complex is unknown. We selected the MCD rat model with prenatal MAM exposure to test the efficacy of mTOR inhibitors in MCDs. We explored the early cortical changes of mTOR pathway protein expression in rats aged P15. We also monitored the early treatment effect of the mTOR inhibitor, rapamycin, on N-methyl-D-aspartate (NMDA)-induced spasms at P15 and their behavior in the juvenile stage. In vivo MR spectroscopy was performed after rapamycin treatment and compared with vehicle controls. There was no difference in mTORC1 pathway protein expression between MAM-exposed MCD rats and controls at P15, and prolonged treatment of rapamycin had no impact on NMDA-induced spasms despite poor weight gain. Prenatal MAM-exposed juvenile rats treated with rapamycin showed increased social approaching and freezing behavior during habituation. MR spectroscopy showed altered neurometabolites, including Gln, Glu+Gln, Tau, and Cr. Despite behavioral changes and in vivo neurometabolic alteration with early prolonged rapamycin treatment, rapamycin had no effect on spasms susceptibility in prenatal MAM-exposed infantile rats with MCD without mTORC1 activation. For MAM-exposed MCD rats without mTORC1 activation, treatment options outside of mTOR pathway inhibitors should be explored.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Eun-Jin Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 05505, Korea
| | - Min-Jee Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea
| | - Mi-Sun Yum
- Department of Pediatrics, University of Ulsan College of Medicine, Seoul, 05505, Korea. .,Department of Pediatrics, Asan Medical Center Children's Hospital, 88 Olympic-ro, Songpa-ku, Seoul, 05505, Korea.
| |
Collapse
|
11
|
Tao X, Zhang Z, Yang Z, Rao B. The effects of taurine supplementation on diabetes mellitus in humans: A systematic review and meta-analysis. FOOD CHEMISTRY. MOLECULAR SCIENCES 2022; 4:100106. [PMID: 35769396 PMCID: PMC9235038 DOI: 10.1016/j.fochms.2022.100106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/19/2022] [Indexed: 12/27/2022]
Abstract
The first meta-analysis to comprehensively evaluate the effects of taurine supplementation on diabetic patients. Statistical significance in HbA1C, Fasting Blood Sugar, HOMA-IR after oral supplemental of taurine by diabetic patients than that of placebo. Taurine is expected to be a new option for the management of diabetes.
Objective The ameliorative effect of taurine on diabetes has received extensive attention in recent years. Despite promising data from animal studies, the efficacy of taurine supplementation in human studies has been inconsistent. We thus did a meta-analysis of randomized controlled trials to assess the effect of taurine supplement on glycemic indices, serum lipids, blood pressure, body composition in patients with diabetes. Methods We systematically searched PubMed, Embase, Cochrane, Web of Science, FDA.gov, and ClinicalTrials.gov for randomized controlled trials (published from inception to January 15, 2022; no language restrictions) about the effect of taurine supplement on diabetes. Values of Standardized Mean Differences (SMD) were determined for continuous outcomes. Results Of 2206 identified studies, 5 randomized controlled trials were eligible and were included in our analysis (N = 209 participants). Compared with the control group, taurine could significantly reduce HbA1c (SMD −0.41[95% CI: −0.74, −0.09], p = 0.01), Fasting Blood Sugar (SMD − 1.28[95% CI: −2.42, −0.14], p = 0.03) and HOMA-IR (SMD − 0.64[95% CI: −1.22, −0.06], p = 0.03). In addition, taurine also reduced Insulin (SMD −0.48 [95% CI: −0.99, 0.03], p = 0.06) and TG (SMD −0.26 [95% CI: −0.55, 0.02], p = 0.07), but did not reach statistical significance. Conclusions Taurine supplementation is beneficial in reducing glycemic indices, such as HbA1c, Fasting Blood Sugar, HOMA-IR in diabetic patients, but has no significant effect on serum lipids, blood pressure and body composition in diabetic patients. Taurine emerges as a new option for the management of patients with diabetes. Further studies are needed to understand the potential effect of taurine in diabetic patients.
Collapse
Affiliation(s)
- Xiaomei Tao
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.,Key Laboratory of Cancer FSMP for State Market Regulation, Beijing 100038, China
| | - Zhanzhi Zhang
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.,Key Laboratory of Cancer FSMP for State Market Regulation, Beijing 100038, China
| | - Zhenpeng Yang
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.,Key Laboratory of Cancer FSMP for State Market Regulation, Beijing 100038, China
| | - Benqiang Rao
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.,Key Laboratory of Cancer FSMP for State Market Regulation, Beijing 100038, China
| |
Collapse
|
12
|
Duszka K. Versatile Triad Alliance: Bile Acid, Taurine and Microbiota. Cells 2022; 11:2337. [PMID: 35954180 PMCID: PMC9367564 DOI: 10.3390/cells11152337] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Taurine is the most abundant free amino acid in the body, and is mainly derived from the diet, but can also be produced endogenously from cysteine. It plays multiple essential roles in the body, including development, energy production, osmoregulation, prevention of oxidative stress, and inflammation. Taurine is also crucial as a molecule used to conjugate bile acids (BAs). In the gastrointestinal tract, BAs deconjugation by enteric bacteria results in high levels of unconjugated BAs and free taurine. Depending on conjugation status and other bacterial modifications, BAs constitute a pool of related but highly diverse molecules, each with different properties concerning solubility and toxicity, capacity to activate or inhibit receptors of BAs, and direct and indirect impact on microbiota and the host, whereas free taurine has a largely protective impact on the host, serves as a source of energy for microbiota, regulates bacterial colonization and defends from pathogens. Several remarkable examples of the interaction between taurine and gut microbiota have recently been described. This review will introduce the necessary background information and lay out the latest discoveries in the interaction of the co-reliant triad of BAs, taurine, and microbiota.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Ramírez-Guerrero S, Guardo-Maya S, Medina-Rincón GJ, Orrego-González EE, Cabezas-Pérez R, González-Reyes RE. Taurine and Astrocytes: A Homeostatic and Neuroprotective Relationship. Front Mol Neurosci 2022; 15:937789. [PMID: 35866158 PMCID: PMC9294388 DOI: 10.3389/fnmol.2022.937789] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 12/20/2022] Open
Abstract
Taurine is considered the most abundant free amino acid in the brain. Even though there are endogenous mechanisms for taurine production in neural cells, an exogenous supply of taurine is required to meet physiological needs. Taurine is required for optimal postnatal brain development; however, its brain concentration decreases with age. Synthesis of taurine in the central nervous system (CNS) occurs predominantly in astrocytes. A metabolic coupling between astrocytes and neurons has been reported, in which astrocytes provide neurons with hypotaurine as a substrate for taurine production. Taurine has antioxidative, osmoregulatory, and anti-inflammatory functions, among other cytoprotective properties. Astrocytes release taurine as a gliotransmitter, promoting both extracellular and intracellular effects in neurons. The extracellular effects include binding to neuronal GABAA and glycine receptors, with subsequent cellular hyperpolarization, and attenuation of N-methyl-D-aspartic acid (NMDA)-mediated glutamate excitotoxicity. Taurine intracellular effects are directed toward calcium homeostatic pathway, reducing calcium overload and thus preventing excitotoxicity, mitochondrial stress, and apoptosis. However, several physiological aspects of taurine remain unclear, such as the existence or not of a specific taurine receptor. Therefore, further research is needed not only in astrocytes and neurons, but also in other glial cells in order to fully comprehend taurine metabolism and function in the brain. Nonetheless, astrocyte’s role in taurine-induced neuroprotective functions should be considered as a promising therapeutic target of several neuroinflammatory, neurodegenerative and psychiatric diseases in the near future. This review provides an overview of the significant relationship between taurine and astrocytes, as well as its homeostatic and neuroprotective role in the nervous system.
Collapse
Affiliation(s)
- Sofía Ramírez-Guerrero
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Santiago Guardo-Maya
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Germán J. Medina-Rincón
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Eduardo E. Orrego-González
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Ricardo Cabezas-Pérez
- Grupo de Investigación en Ciencias Biomédicas GRINCIBIO, Facultad de Medicina, Universidad Antonio Nariño, Bogotá, Colombia
| | - Rodrigo E. González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
- *Correspondence: Rodrigo E. González-Reyes,
| |
Collapse
|
14
|
Khalaf HA, Elsamanoudy AZ, Abo-Elkhair SM, Hassan FE, Mohie PM, Ghoneim FM. Endoplasmic reticulum stress and mitochondrial injury are critical molecular drivers of AlCl 3-induced testicular and epididymal distortion and dysfunction: protective role of taurine. Histochem Cell Biol 2022; 158:97-121. [PMID: 35511291 PMCID: PMC9247002 DOI: 10.1007/s00418-022-02111-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2022] [Indexed: 11/30/2022]
Abstract
Aluminum, the third most plentiful metal in the Earth's crust, has potential for human exposure and harm. Oxidative stress plays an essential role in producing male infertility by inducing defects in sperm functions. We aimed to investigate the role of endoplasmic reticulum (ER) stress and mitochondrial injury in the pathogenesis of aluminum chloride (AlCl3)-induced testicular and epididymal damage at the histological, biochemical, and molecular levels, and to assess the potential protective role of taurine. Forty-eight adult male albino rats were separated into four groups (12 in each): negative control, positive control, AlCl3, and AlCl3 plus taurine groups. Testes and epididymis were dissected. Histological and immunohistochemical (Bax and vimentin) studies were carried out. Gene expression of vimentin, PCNA, CHOP, Bcl-2, Bax, and XBP1 were investigated via quantitative real-time polymerase chain reaction (qRT-PCR), besides estimation of malondialdehyde (MDA) and total antioxidant capacity (TAC). Light and electron microscopic examinations of the testes and epididymis revealed pathological changes emphasizing both mitochondrial injury and ER stress in the AlCl3 group. Taurine-treated rats showed a noticeable improvement in the testicular and epididymal ultrastructure. Moreover, they exhibited increased gene expression of vimentin, Bcl-2, and PNCA accompanied by decreased CHOP, Bax, and XBP1 gene expression. In conclusion, male reproductive impairment is a significant hazard associated with AlCl3 exposure. Both ER stress and mitochondrial impairment are critical mechanisms of the deterioration in the testes and epididymis induced by AlCl3, but taurine can amend this.
Collapse
Affiliation(s)
- Hanaa A Khalaf
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Ayman Z Elsamanoudy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah, 21465, Saudi Arabia
| | - Salwa M Abo-Elkhair
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Passant M Mohie
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fatma M Ghoneim
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
15
|
Oja SS, Saransaari P. Taurine and the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:325-331. [DOI: 10.1007/978-3-030-93337-1_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
16
|
Posey EA, Bazer FW, Wu G. Amino Acids and Their Metabolites for Improving Human Exercising Performance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1332:151-166. [PMID: 34251643 DOI: 10.1007/978-3-030-74180-8_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Achieving adequate nutrition for exercising humans is especially important for improving both muscle mass and metabolic health. One of the most common misunderstandings in the fitness industry is that the human body has requirements for dietary whole protein and that exercising individuals must consume only whole protein to meet their physiological needs. This view, however, is incorrect. Instead, humans at rest or during exercise have requirements for dietary amino acids (AAs), and dietary protein is a source of AAs in the body. The requirements for AAs must be met each day to avoid a negative nitrogen balance in individuals with moderate or intense physical activity. By properly meeting increased requirements for AAs through increased intake of high-quality protein (the source of AAs) plus supplemental AAs, athletes can improve their overall athletic performance. AAs or metabolites that are of special importance for exercising individuals include arginine, branched-chain AAs, creatine, glycine, taurine, and glutamine. The AAs play vital roles as both substrates for protein synthesis and molecules for regulating blood flow and nutrient metabolism. The functional roles of AAs include the maintenance of cell and tissue integrity; stimulation of mechanistic target of rapamycin and AMP-activated protein kinase cell signaling pathways; energy sources for the small intestine, cells of the immune system, and skeletal muscle; antioxidant and anti-inflammatory reactions; production of neurotransmitters; modulation of acid-base balance in the body. All of those roles are crucial for the overall goal of improving exercise performance. Therefore, adequate intakes of proteinogenic AAs and their functional metabolites, especially those noted in this review, are essential for optimal human health (including optimum muscle mass and function) and should be a primary goal of exercising individuals.
Collapse
Affiliation(s)
- Erin A Posey
- Department of Animal Science, Texas A&M University, College Station, Texas, TX, 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, TX, 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, Texas, TX, 77843, USA.
| |
Collapse
|
17
|
Kumar S, Goel RK. Pharmacokinetic, pharmacodynamic, and neurochemical investigations of lamotrigine-pentylenetetrazole kindled mice to ascertain it as a reliable model for clinical drug-resistant epilepsy. Animal Model Exp Med 2020; 3:245-255. [PMID: 33024946 PMCID: PMC7529331 DOI: 10.1002/ame2.12131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Pentylenetetrazole kindling has long been used for the screening of investigational antiseizure drugs. The presence of lamotrigine, at a very low dose, does not hamper kindling in mice; rather it modifies this epileptogenesis process into drug-resistant epilepsy. The lamotrigine-pentylenetetrazole kindled mice show resistance to lamotrigine, phenytoin, and carbamazepine. It may also be possible that other licensed antiseizure drugs, like the mentioned drugs, remain ineffective in this model; therefore, this was the subject of this study. METHODS Swiss albino mice were kindled with pentylenetetrazole for 35 days in the presence of either methylcellulose vehicle or lamotrigine (subtherapeutic dose, ie, 5 mg/kg). Vehicle vs lamotrigine-kindled mice were compared in terms of (a) resistance/response toward nine antiseizure drugs applied as monotherapies and two drug combinations; (b) lamotrigine bioavailability in blood and brain; (c) blood-brain barrier integrity; and (d) amino acids and monoamines in the cerebral cortex and hippocampus. RESULTS Lamotrigine vs vehicle-kindled mice are similar (or not significantly different P > .05 from each other) in terms of (a) response toward drug combinations; (b) lamotrigine bioavailability; and (c) blood-brain barrier integrity except for, significantly (P < .05) reduced taurine and increased glutamate in the cerebral cortex and hippocampus. Aside from these, lamotrigine-kindled mice show significant (P < .05) resistant to lamotrigine (15 mg/kg), levetiracetam (40 mg/kg); carbamazepine (40 mg/kg), zonisamide (100 mg/kg), gabapentin (224 mg/kg), pregabalin (30 mg/kg), phenytoin (35 mg/kg), and topiramate (300 mg/kg). CONCLUSION Lamotrigine-pentylenetetrazole kindling takes longer to develop (~5 weeks) in comparison to lamotrigine-amygdale (~4 weeks) and lamotrigine-corneal (~2 weeks) kindling models. However, drug screening through this model may yield superior drugs with novel antiseizure mechanisms.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Pharmaceutical Sciences & Drug Research Punjabi University Patiala Punjab India
| | - Rajesh K Goel
- Department of Pharmaceutical Sciences & Drug Research Punjabi University Patiala Punjab India
| |
Collapse
|
18
|
Francisco EDS, Mendes-da-Silva RF, de Castro CBL, Soares GDSF, Guedes RCA. Taurine/Pilocarpine Interaction in the Malnourished Rat Brain: A Behavioral, Electrophysiological, and Immunohistochemical Analysis. Front Neurosci 2019; 13:981. [PMID: 31619952 PMCID: PMC6759493 DOI: 10.3389/fnins.2019.00981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/30/2019] [Indexed: 12/05/2022] Open
Abstract
This study aimed to evaluate the possible protective role of taurine on anxiety-like behavior, brain electrical activity and glial cell immunoreactivity in well-nourished and malnourished rats that were treated with a subconvulsing dose of pilocarpine. Newborn Wistar rats were subjected to normal or unfavorable lactation conditions, represented by the suckling of litters with 9 or 15 pups, resulting in well-nourished and malnourished animals, respectively. Each nutritional group was split into five subgroups that were treated from postnatal day (PND) 35 to 55 with 300 mg/kg/day of taurine + 45 mg/kg/day of pilocarpine (group T + P), taurine only (group T), pilocarpine only (group P), vehicle control (group V), or not treated control (group naïve; Nv). At PND56-58, the groups were subjected to the elevated plus-maze behavioral tests. Glycemia was measured on PND59. Between PND60 and PND65, the cortical spreading depression (CSD) was recorded in the cerebral cortex, and the levels of malondialdehyde and microglial and astrocyte immunoreactivity were evaluated in the cortex and hippocampus. Our data indicate that treatment with taurine and pilocarpine resulted in anxiolytic-like and anxiogenic behavior, respectively, and that nutritional deficiency modulated these effects. Both treatments decelerated CSD propagation and modulated GFAP- and Iba1-containing glial cells. Pilocarpine reduced body weight and glycemia, and administration of taurine was not able to attenuate the effects of pilocarpine. The molecular mechanisms underlying taurine action on behavioral and electrophysiological parameters in the normal and altered brain remain to be further explored.
Collapse
|
19
|
Bertoncello KT, Müller TE, Fontana BD, Franscescon F, Filho GLB, Rosemberg DB. Taurine prevents memory consolidation deficits in a novel alcohol-induced blackout model in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:39-45. [PMID: 30880191 DOI: 10.1016/j.pnpbp.2019.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023]
Abstract
Ethanol is one of the most consumed substance worldwide that impairs learning and memory processes, resulting in amnesia or blackout. Due to the genetic conservation, rich behavioral repertoire, and high pharmacological tractability, the zebrafish (Danio rerio) has emerged as a powerful model organism for assessing preventive strategies against the noxious effects of ethanol in vertebrates. Here, we used an inhibitory avoidance apparatus to investigate the potential preventive effects of taurine in a novel ethanol-induced amnesia model in zebrafish. The experimental tank consisted of two compartments of the same size, one dark and another white, which were separated by a guillotine-type door. Three parallel metal bars coupled to an electrical stimulator were connected on each lateral wall of the dark compartment as electrical stimulus source. Differences on the latency to enter the dark compartment were used as retention indexes. A mild electric shock (125 mA, 3 ± 0.2 V) at 10 and 1000 Hz did not promote significant learning, while 100 Hz facilitated memory retention. Posttraining administration of MK-801 blocked this response, reinforcing the predictive validity of the test. Treatments were performed immediately after the training session using the 100 Hz frequency. Animals were exposed to water (control), taurine (42, 150, 400 mg/L), ethanol (0.25%, 1.0% v/v) or taurine plus ethanol to assess the effects on memory consolidation. Test session was performed 24 h following training. Ethanol at 0.25% did not affect memory consolidation, but 1.0% impaired memory without changing locomotion. Although taurine alone did not modulate learning, all concentrations tested exerted prevented ethanol-induced memory impairment. Overall, we describe a novel ethanol-induced blackout model, where a high ethanol concentration acutely impairs memory consolidation in zebrafish. Moreover, since taurine showed a protective role, we reinforce the growing utility of zebrafish models for assessing the deleterious effects of ethanol and potential therapeutic strategies.
Collapse
Affiliation(s)
- Kanandra T Bertoncello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| | - Talise E Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Barbara D Fontana
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Francini Franscescon
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Gilvan L B Filho
- Department of Biomedical Equipment. Federal Institute of Education, Science and Technology. s/n BR 406, Km 145. Ceará-Mirim, RN 59570-000, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
20
|
Abstract
Malnutrition is a common feature of chronic and acute diseases, often associated with a poor prognosis, including worsening of clinical outcome, owing, among other factors, to dysfunction of the most internal organs and systems affecting the absorption, metabolism and elimination of drugs and nutrients. Taurine is involved in numerous biological processes and is required in increased amounts in response to pathological conditions. The aim of this study was to describe the behaviour of taurine in well-nourished (WN) rats and to analyse the influence of protein-energy undernutrition on the pharmacokinetic (PK) parameters of taurine, using a PK model. Wistar rats were randomly distributed into two groups, WN and undernourished (UN), and taurine was administered intravenously or orally at different doses: 1, 10 and 100 mg. Population pharmacokinetic modelling of plasma levels was performed using the NONMEM 7.2 program. Several distribution and absorption models were explored in combination with dose and/or time covariate effects. Covariates such as nutritional status, serum albumin, body weight and score of undernutrition were used. A two-compartment population pharmacokinetic model with zero-order endogenous formation, passive absorption, first-order kinetics distribution and non-linear elimination with parallel Michaelis-Menten excretion and reabsorption processes best described taurine pharmacokinetics. Undernutrition acted as a covariate reducing the V max of the active elimination process. Data analysis showed linear absorption and distribution, and non-linear elimination processes for taurine. Elimination of taurine was reduced in UN animals, suggesting that the reabsorption process via the secretion transporter was modified in that group.
Collapse
|
21
|
Gupte R, Christian S, Keselman P, Habiger J, Brooks WM, Harris JL. Evaluation of taurine neuroprotection in aged rats with traumatic brain injury. Brain Imaging Behav 2019; 13:461-471. [PMID: 29656312 PMCID: PMC6186512 DOI: 10.1007/s11682-018-9865-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Despite higher rates of hospitalization and mortality following traumatic brain injury (TBI) in patients over 65 years old, older patients remain underrepresented in drug development studies. Worse outcomes in older individuals compared to younger adults could be attributed to exacerbated injury mechanisms including oxidative stress, inflammation, blood-brain barrier disruption, and bioenergetic dysfunction. Accordingly, pleiotropic treatments are attractive candidates for neuroprotection. Taurine, an endogenous amino acid with antioxidant, anti-inflammatory, anti-apoptotic, osmolytic, and neuromodulator effects, is neuroprotective in adult rats with TBI. However, its effects in the aged brain have not been evaluated. We subjected aged male rats to a unilateral controlled cortical impact injury to the sensorimotor cortex, and randomized them into four treatment groups: saline or 25 mg/kg, 50 mg/kg, or 200 mg/kg i.p. taurine. Treatments were administered 20 min post-injury and daily for 7 days. We assessed sensorimotor function on post-TBI days 1-14 and tissue loss on day 14 using T2-weighted magnetic resonance imaging. Experimenters were blinded to the treatment group for the duration of the study. We did not observe neuroprotective effects of taurine on functional impairment or tissue loss in aged rats after TBI. These findings in aged rats are in contrast to previous reports of taurine neuroprotection in younger animals. Advanced age is an important variable for drug development studies in TBI, and further research is required to better understand how aging may influence mechanisms of taurine neuroprotection.
Collapse
Affiliation(s)
- Raeesa Gupte
- Hoglund Brain Imaging Center, University of Kansas Medical Center, KS 66160, USA, 913-588-3519,
| | - Sarah Christian
- Hoglund Brain Imaging Center, University of Kansas Medical Center, KS 66160, USA, 913-588-9070,
| | - Paul Keselman
- Hoglund Brain Imaging Center, University of Kansas Medical Center, KS 66160, USA, 913-588-9079,
| | - Joshua Habiger
- Department of Biostatistics, University of Kansas Medical Center, KS 66160, USA, 405-744-9657,
| | - William M. Brooks
- Department of Neurology, Director, Hoglund Brain Imaging Center, Director, University of Kansas Alzheimer’s Disease Center Neuroimaging Core, University of Kansas Medical Center, KS 66160, USA, 913-588-9075,
| | - Janna L. Harris
- Department of Anatomy & Cell Biology, Director, Animal Magnetic Resonance Imaging Core, Hoglund Brain Imaging Center, University of Kansas Medical Center, KS 66160, USA, 913-588-9076,
| |
Collapse
|
22
|
Ochoa-de la Paz L, Zenteno E, Gulias-Cañizo R, Quiroz-Mercado H. Taurine and GABA neurotransmitter receptors, a relationship with therapeutic potential? Expert Rev Neurother 2019; 19:289-291. [DOI: 10.1080/14737175.2019.1593827] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Lenin Ochoa-de la Paz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
- Departamento de Investigación, Asociación para Evitar la Ceguera en México I.A.P. Hospital Dr. Luis Sánchez Bulnes, México City, México
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | - Rosario Gulias-Cañizo
- Departamento de Investigación, Asociación para Evitar la Ceguera en México I.A.P. Hospital Dr. Luis Sánchez Bulnes, México City, México
| | - Hugo Quiroz-Mercado
- Departamento de Investigación, Asociación para Evitar la Ceguera en México I.A.P. Hospital Dr. Luis Sánchez Bulnes, México City, México
| |
Collapse
|
23
|
Winkler P, Luhmann HJ, Kilb W. Taurine potentiates the anticonvulsive effect of the GABA A agonist muscimol and pentobarbital in the immature mouse hippocampus. Epilepsia 2019; 60:464-474. [PMID: 30682212 DOI: 10.1111/epi.14651] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The high incidence of epileptic seizures in neonates and their frequent refractoriness to pharmacologic therapies require identification of new therapeutical options. Therefore, we investigated whether the modulatory effect of taurine on γ-aminobutyric acid (GABA)A receptors can enhance the anticonvulsive potential of the GABAA receptor agonist muscimol and of the barbiturate pentobarbital. METHODS We performed field potential recordings in in toto hippocampus preparations of immature (postnatal days 4-7) C57Bl/6 mouse pups. Spontaneous epileptiform activity was induced by the continuous presence of the potassium channel blocker 4-aminopyridine and the glycinergic antagonist strychnine in Mg2+ -free solutions. RESULTS Bath application of 0.1 μmol/L muscimol increases the occurrence of recurrent epileptiform discharges, whereas they are significantly attenuated in a dose-dependent manner by muscimol in concentrations between 0.5 and 5 μmol/L. Taurine at concentrations between 0.1 and 0.5 mmol/L induces a proconvulsive effect, but upon coapplication, it significantly augments the anticonvulsive effect of moderate muscimol doses (0.5-1 μmol/L). In addition, the anticonvulsive effect of 100 and 200 μmol/L pentobarbital is increased significantly in the presence of 0.5 μmol/L taurine. SIGNIFICANCE These observations demonstrate that taurine can indeed enhance the anticonvulsive effects of muscimol and pentobarbital, suggesting that taurine may act as a positive modulator on GABAA receptors. Thus, interfering with the modulatory taurine binding site of GABAA receptors or the interstitial taurine concentration may provide new therapeutical options for anticonvulsive therapies in neonates.
Collapse
Affiliation(s)
- Paula Winkler
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
24
|
Mitochondria protecting amino acids: Application against a wide range of mitochondria-linked complications. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
25
|
Kilb W. Putative Role of Taurine as Neurotransmitter During Perinatal Cortical Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:281-292. [PMID: 28849463 DOI: 10.1007/978-94-024-1079-2_25] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neurotransmitters and neuronal activity affect neurodevelopmental events like neurogenesis, neuronal migration, apoptosis and differentiation. Beside glutamate and gamma-amino butyric acid, the aminosulfonic acid taurine has been considered as possible neurotransmitter that influences early neuronal development. In this article I review recent studies of our group which demonstrate that taurine can affect a variety of identified neuronal populations in the immature neocortex and directly modulates neuronal activity. These experiments revealed that taurine evoke dose-dependent membrane responses in a variety of neocortical neuron populations, including Cajal-Retzius cells, subplate neurons and GABAergic interneurons. Taurine responses persist in the presence of GABA(A) receptor antagonists and are reduced by the addition of strychnine, suggesting that glycine receptors are involved in taurine-mediated membrane responses. Gramicidin-perforated patch-clamp and cell-attached recordings demonstrated that taurine evokes depolarizing and mainly excitatory membrane responses, in accordance with the high intracellular Cl- concentration in immature neurons. In addition, taurine increases the frequency of postsynaptic GABAergic currents (PSCs) in a considerable fraction of immature pyramidal neurons, indicating a specific activation of presynaptic GABAergic networks projecting toward and exciting pyramidal neurons. In summary, these results suggest that taurine may be critically involved in the regulation of network excitability in the immature neocortex and hippocampus via interactions with glycine receptors.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology, University Medical Center, Johannes-Gutenberg University, Mainz, Germany.
| |
Collapse
|
26
|
Schaffer S, Kim HW. Effects and Mechanisms of Taurine as a Therapeutic Agent. Biomol Ther (Seoul) 2018; 26:225-241. [PMID: 29631391 PMCID: PMC5933890 DOI: 10.4062/biomolther.2017.251] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 01/16/2023] Open
Abstract
Taurine is an abundant, β-amino acid with diverse cytoprotective activity. In some species, taurine is an essential nutrient but in man it is considered a semi-essential nutrient, although cells lacking taurine show major pathology. These findings have spurred interest in the potential use of taurine as a therapeutic agent. The discovery that taurine is an effective therapy against congestive heart failure led to the study of taurine as a therapeutic agent against other disease conditions. Today, taurine has been approved for the treatment of congestive heart failure in Japan and shows promise in the treatment of several other diseases. The present review summarizes studies supporting a role of taurine in the treatment of diseases of muscle, the central nervous system, and the cardiovascular system. In addition, taurine is extremely effective in the treatment of the mitochondrial disease, mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), and offers a new approach for the treatment of metabolic diseases, such as diabetes, and inflammatory diseases, such as arthritis. The review also addresses the functions of taurine (regulation of antioxidation, energy metabolism, gene expression, ER stress, neuromodulation, quality control and calcium homeostasis) underlying these therapeutic actions.
Collapse
Affiliation(s)
- Stephen Schaffer
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688,
USA
| | - Ha Won Kim
- Department of Life Science, University of Seoul, Seoul 02504,
Republic of Korea
| |
Collapse
|
27
|
Nedergaard S, Andreasen M. Opposing effects of 2-deoxy-d-glucose on interictal- and ictal-like activity when K+ currents and GABAA receptors are blocked in rat hippocampus in vitro. J Neurophysiol 2018; 119:1912-1923. [DOI: 10.1152/jn.00732.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ketogenic diet (KD), a high-fat, carbohydrate-restricted diet, is used as an alternative treatment for drug-resistant epileptic patients. Evidence suggests that compromised glucose metabolism has a significant role in the anticonvulsant action of the KD; however, it is unclear what part of the glucose metabolism that is important. The present study investigates how selective alterations in glycolysis and oxidative phosphorylation influence epileptiform activity induced by blocking K+ currents and GABAA and NMDA receptors in the hippocampal slice preparation. Blocking glycolysis with the glucose derivative 2-deoxy-d-glucose (2-DG; 10 mM) gave a fast reduction of the frequency of interictal discharge (IED) consistent with findings in other in vitro models. However, this was followed by the induction of seizure-like discharges in area CA1 and CA3. Substituting glucose with sucrose (glucopenia) had effects similar to those of 2-DG, whereas substitution with l-lactate or pyruvate reduced the IED but had a less proconvulsant effect. Blockade of ATP-sensitive K+ channels, glycine or adenosine 1 receptors, or depletion of the endogenous anticonvulsant compound glutathione did not prevent the actions of 2-DG. Baclofen (2 μM) reproduced the effect of 2-DG on IED activity. The proconvulsant effect of 2-DG could be reproduced by blocking the oxidative phosphorylation with the complex I toxin rotenone (4 μM). The data suggest that inhibition of IED, induced by 2-DG and glucopenia, is a direct consequence of impairment of glycolysis, likely exerted via a decreased recurrent excitatory synaptic transmission in area CA3. The accompanying proconvulsant effect is caused by an excitatory mechanism, depending on impairment of oxidative phosphorylation. NEW & NOTEWORTHY This study reveals two opposing effects of 2-deoxy-d-glucose (2-DG) and glucopenia on in vitro epileptiform discharge observed during combined blockade of K+ currents and GABAA receptors. Interictal-like activity is inhibited by a mechanism that selectively depends on impairment of glycolysis and that results from a decrease in the strength of excitatory recurrent synaptic transmission in area CA3. In contrast, 2-DG and glucopenia facilitate ictal-like activity by an excitatory mechanism, depending on impairment of mitochondrial oxidative phosphorylation.
Collapse
|
28
|
Effect of levetiracetam on extracellular amino acid levels in the dorsal hippocampus of rats with temporal lobe epilepsy. Epilepsy Res 2018; 140:111-119. [PMID: 29331845 DOI: 10.1016/j.eplepsyres.2018.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/08/2017] [Accepted: 01/03/2018] [Indexed: 12/23/2022]
Abstract
Levetiracetam (LEV) is an anticonvulsant drug with a unique mechanism of action that is not completely understood. However, its activity profile may involve effects on excitatory and/or inhibitory neurotransmission since the primary target of LEV, synaptic vesicle protein 2A, is ubiquitously expressed in all types of synaptic vesicles. Therefore, the objective of the present study was to explore the effect of LEV (300 mg/kg/day for one week, administered via osmotic mini-pumps) on neurotransmitter release and its probable selective effect on extracellular gamma-amino butyric acid (GABA), glutamate (Glu), aspartate (Asp), glutamine (Gln), taurine (Tau) and glycine (Gly) concentrations (using in vivo microdialysis under basal and high-K+ conditions) in the dorsal hippocampus (DH), a region that undergoes major synaptic changes during epilepsy. Epileptic rats developed clear signs of hyperexcitability, i.e., an elevated Glu/GABA ratio in the DH. The LEV concentration in blood after 7 days of treatment was within the therapeutic range. In contrast, LEV was not detected four days after mini-pump removal (washout period). Furthermore, LEV restored the Glu/GABA ratio to approximately the control level and significantly increased the GABA concentration after the initiation of high-K+ conditions. Based on these data, LEV treatment restored the lost balance between the excitatory and inhibitory systems under basal conditions. Moreover, LEV showed a selective effect by preferentially increasing vesicular release of GABA, a mechanism by which LEV could reduce epileptic seizures.
Collapse
|
29
|
Kilb W, Fukuda A. Taurine as an Essential Neuromodulator during Perinatal Cortical Development. Front Cell Neurosci 2017; 11:328. [PMID: 29123472 PMCID: PMC5662885 DOI: 10.3389/fncel.2017.00328] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/04/2017] [Indexed: 01/10/2023] Open
Abstract
A variety of experimental studies demonstrated that neurotransmitters are an important factor for the development of the central nervous system, affecting neurodevelopmental events like neurogenesis, neuronal migration, programmed cell death, and differentiation. While the role of the classical neurotransmitters glutamate and gamma-aminobutyric acid (GABA) on neuronal development is well established, the aminosulfonic acid taurine has also been considered as possible neuromodulator during early neuronal development. The purpose of the present review article is to summarize the properties of taurine as neuromodulator in detail, focusing on the direct involvement of taurine on various neurodevelopmental events and the regulation of neuronal activity during early developmental epochs. The current knowledge is that taurine lacks a synaptic release mechanism but is released by volume-sensitive organic anion channels and/or a reversal of the taurine transporter. Extracellular taurine affects neurons and neuronal progenitor cells mainly via glycine, GABA(A), and GABA(B) receptors with considerable receptor and subtype-specific affinities. Taurine has been shown to directly influence neurogenesis in vitro as well as neuronal migration in vitro and in vivo. It provides a depolarizing signal for a variety of neuronal population in the immature central nervous system, thereby directly influencing neuronal activity. While in the neocortex, taurine probably enhance neuronal activity, in the immature hippocampus, a tonic taurinergic tone might be necessary to attenuate activity. In summary, taurine must be considered as an essential modulator of neurodevelopmental events, and possible adverse consequences on fetal and/or early postnatal development should be evaluated for pharmacological therapies affecting taurinergic functions.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology, University Medical Center, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
30
|
Kaur N, Singh T, Kumar S, Goel RK. Neurochemical evidence based suggested therapy for safe management of epileptogenesis. Epilepsy Behav 2017; 72:8-16. [PMID: 28570965 DOI: 10.1016/j.yebeh.2017.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/03/2017] [Indexed: 01/05/2023]
Abstract
Most of the clinically available antiepileptic drugs have only antiseizure effects and are reported unable to prevent epileptogenesis. In the past decade, several drugs underwent clinical trials for management of epileptogenesis, but none of the drugs tested was found effective. One of the major lacunas is availability of appropriate preclinical approaches to delineate mechanisms of epileptogenesis. Thus, the present study attempts to suggest a neurochemistry based approach for safe management of epileptogenesis. The altered neurochemical milieu in amygdala, cortex and hippocampus areas of the mice brain in naïve, kindled and kindling resistant animals has been delineated. The endogenous natural antiepileptogenic neurochemical defense mechanism observed in kindling resistant animals may uncover neurochemical mechanisms of epileptogenesis and in turn suggest us novel interventions for safe management of epileptogenesis. The kindling epileptogenesis was carried out in two month old male Swiss albino mice by administering subconvulsive pentylenetetrazole (35mg/kg; i.p.) at an interval of 48±2h for 42days. 2h after the last pentylenetetrazole injection, the animals were subjected to behavioral evaluations. Four hours after behavioral evaluation, all animals were euthanized and discrete parts of brain (amygdala, cortex and hippocampus) were harvested for neurochemical analysis. Results revealed that 60% of animals responded to kindling as observed with decreased seizure threshold, while the rest were found resistant. The kindled animals were found to be associated with anxiety, depression and cognitive impairment; while in kindling resistant animals no such behavioral deficits were observed. The neurochemical analysis revealed that in kindled animals altered glutamate-GABA neurotransmission, and decreased taurine, glycine, d-serine, monoamine levels with elevated indoleamine 2,3-dioxygenase activity were observed, which may be convicted for progression of kindling epileptogenesis. However, in kindling resistant animals elevated GABA, taurine, tryptophan, serotonin, glycine, and d-serine levels with decreased indoleamine 2,3-dioxygenase activity were observed as natural endogenous antiepileptogenic mechanisms, which may be foreseen as safe pharmacological targets for management of epileptogenesis.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Tanveer Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Sandeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Rajesh Kumar Goel
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India.
| |
Collapse
|
31
|
Glick NR, Fischer MH. Potential Benefits of Ameliorating Metabolic and Nutritional Abnormalities in People With Profound Developmental Disabilities. Nutr Metab Insights 2017; 10:1178638817716457. [PMID: 35185339 PMCID: PMC8855413 DOI: 10.1177/1178638817716457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/21/2017] [Indexed: 11/20/2022] Open
Abstract
Background: People with profound developmental disabilities have some of the most severe neurological impairments seen in society, have accelerated mortality due to huge medical challenges, and yet are often excluded from scientific studies. They actually have at least 2 layers of conditions: (1) the original disability and (2) multiple under-recognized and underexplored metabolic and nutritional imbalances involving minerals (calcium, zinc, and selenium), amino acids (taurine, tryptophan), fatty acids (linoleic acid, docosahexaenoic acid, arachidonic acid, adrenic acid, Mead acid, plasmalogens), carnitine, hormones (insulinlike growth factor 1), measures of oxidative stress, and likely other substances and systems. Summary: This review provides the first list of metabolic and nutritional abnormalities commonly found in people with profound developmental disabilities and, based on the quality of life effects of similar abnormalities in neurotypical people, indicates the potential effects of these abnormalities in this population which often cannot communicate symptoms. Key messages: We propose that improved understanding and management of these disturbed mechanisms would enhance the quality of life of people with profound developmental disabilities. Such insights may also apply to people with other conditions associated with disability, including some diseases requiring stem cell implantation and living in microgravity.
Collapse
Affiliation(s)
- Norris R Glick
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Milton H Fischer
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
32
|
Ochiai Y, Uchida Y, Ohtsuki S, Tachikawa M, Aizawa S, Terasaki T. The blood-brain barrier fatty acid transport protein 1 (FATP1/SLC27A1) supplies docosahexaenoic acid to the brain, and insulin facilitates transport. J Neurochem 2017; 141:400-412. [PMID: 28035674 DOI: 10.1111/jnc.13943] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 12/14/2016] [Accepted: 12/14/2016] [Indexed: 11/28/2022]
Abstract
We purposed to clarify the contribution of fatty acid transport protein 1 (FATP1/SLC 27A1) to the supply of docosahexaenoic acid (DHA) to the brain across the blood-brain barrier in this study. Transport experiments showed that the uptake rate of [14 C]-DHA in human FATP1-expressing HEK293 cells was significantly greater than that in empty vector-transfected (mock) HEK293 cells. The steady-state intracellular DHA concentration was nearly 2-fold smaller in FATP1-expressing than in mock cells, suggesting that FATP1 works as not only an influx, but also an efflux transporter for DHA. [14 C]-DHA uptake by a human cerebral microvascular endothelial cell line (hCMEC/D3) increased in a time-dependent manner, and was inhibited by unlabeled DHA and a known FATP1 substrate, oleic acid. Knock-down of FATP1 in hCMEC/D3 cells with specific siRNA showed that FATP1-mediated uptake accounts for 59.2-73.0% of total [14 C]-DHA uptake by the cells. Insulin treatment for 30 min induced translocation of FATP1 protein to the plasma membrane in hCMEC/D3 cells and enhanced [14 C]-DHA uptake. Immunohistochemical analysis of mouse brain sections showed that FATP1 protein is preferentially localized at the basal membrane of brain microvessel endothelial cells. We found that two neuroprotective substances, taurine and biotin, in addition to DHA, undergo FATP1-mediated efflux. Overall, our results suggest that FATP1 localized at the basal membrane of brain microvessels contributes to the transport of DHA, taurine and biotin into the brain, and insulin rapidly increases DHA supply to the brain by promoting translocation of FATP1 to the membrane. Read the Editorial Comment for this article on page 324.
Collapse
Affiliation(s)
- Yusuke Ochiai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Japan Agency for Medical Research and Development (AMED) CREST, Tokyo, Japan
| | - Masanori Tachikawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Sanshiro Aizawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| |
Collapse
|
33
|
Morland C, Pettersen MN, Hassel B. Hyperosmolar sodium chloride is toxic to cultured neurons and causes reduction of glucose metabolism and ATP levels, an increase in glutamate uptake, and a reduction in cytosolic calcium. Neurotoxicology 2016; 54:34-43. [PMID: 26994581 DOI: 10.1016/j.neuro.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 02/01/2023]
Abstract
Elevation of serum sodium, hypernatremia, which may occur during dehydration or treatment with sodium chloride, may cause brain dysfunction and damage, but toxic mechanisms are poorly understood. We found that exposure to excess NaCl, 10-100mmol/L, for 20h caused cell death in cultured cerebellar granule cells (neurons). Toxicity was due to Na(+), since substituting excess Na(+) with choline reduced cell death to control levels, whereas gluconate instead of excess Cl(-) did not. Prior to cell death from hyperosmolar NaCl, glucose consumption and lactate formation were reduced, and intracellular aspartate levels were elevated, consistent with reduced glycolysis or glucose uptake. Concomitantly, the level of ATP became reduced. Pyruvate, 10mmol/L, reduced NaCl-induced cell death. The extracellular levels of glutamate, taurine, and GABA were concentration-dependently reduced by excess NaCl; high-affinity glutamate uptake increased. High extracellular [Na(+)] caused reduction in intracellular free [Ca(2+)], but a similar effect was seen with mannitol, which was not neurotoxic. We suggest that inhibition of glucose metabolism with ensuing loss of ATP is a neurotoxic mechanism of hyperosmolar sodium, whereas increased uptake of extracellular neuroactive amino acids and reduced intracellular [Ca(2+)] may, if they occur in vivo, contribute to the cerebral dysfunction and delirium described in hypernatremia.
Collapse
Affiliation(s)
- Cecilie Morland
- Norwegian Defence Research Establishment, Kjeller, Norway; Oslo and Akershus University College of Applied Sciences, Oslo, Norway
| | | | - Bjørnar Hassel
- Norwegian Defence Research Establishment, Kjeller, Norway; Department of Complex Neurology and Neurohabilitation, Oslo University Hospital and The University of Oslo, Oslo, Norway.
| |
Collapse
|
34
|
Heidari R, Rasti M, Shirazi Yeganeh B, Niknahad H, Saeedi A, Najibi A. Sulfasalazine-induced renal and hepatic injury in rats and the protective role of taurine. BIOIMPACTS : BI 2016; 6:3-8. [PMID: 27340618 PMCID: PMC4916549 DOI: 10.15171/bi.2016.01] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 12/19/2015] [Accepted: 12/26/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Sulfasalazine is a drug commonly administrated against inflammatory-based disorders. On the other hand, kidney and liver injury are serious adverse events accompanied by sulfasalazine administration. No specific therapeutic option is available against this complication. The current investigation was designed to evaluate the potential protective effects of taurine against sulfasalazine-induced kidney and liver injury in rats. METHODS Male Sprague-Dawley rats were administered with sulfasalazine (600 mg/kg, oral) for 14 consecutive days. Animals received different doses of taurine (250, 500 and 1000 mg/ kg, i.p.) every day. Markers of organ injury were evaluated on day 15(th), 24 h after the last dose of sulfasalazine. RESULTS Sulfasalazine caused renal and hepatic injury as judged by an increase in serum level of creatinine (Cr), alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP). The levels of reactive oxygen species (ROS) and lipid peroxidation were raised in kidney and liver of sulfasalazine-treated animals. Moreover, tissue glutathione reservoirs were depleted after sulfasalazine administration. Histopathological changes of kidney and liver also endorsed organ injury. Taurine administration (250, 500 and 1000 mg/kg/day, i.p) alleviated sulfasalazine-induced renal and hepatic damage. CONCLUSION Taurine administration could serve as a potential protective agent with therapeutic capabilities against sulfasalazine adverse effects.
Collapse
Affiliation(s)
- Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Rasti
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Babak Shirazi Yeganeh
- Department of Pathology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Corresponding author: Hossein Niknahad,
| | - Arastoo Saeedi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Najibi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
35
|
Efficacy of the Ketogenic Diet for the Treatment of Refractory Childhood Epilepsy: Cerebrospinal Fluid Neurotransmitters and Amino Acid Levels. Pediatr Neurol 2015; 53:422-6. [PMID: 26476148 DOI: 10.1016/j.pediatrneurol.2015.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/21/2015] [Accepted: 07/26/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The mechanisms of the ketogenic diet remain unclear, but several predictors of response have been proposed. We aimed is to study the relationship between the etiology of epilepsy, cerebrospinal fluid neurotransmitters, pterins, and amino acids, and response to a ketogenic diet. METHODS We studied 60 patients who began classic ketogenic diet treatment for refractory epilepsy. In 24 of 60 individuals, we analyzed cerebrospinal fluid neurotransmitters, pterins, and amino acids in baseline conditions. Mean age at epilepsy onset was 24 months, 83.3% were focal epilepsies, and in 51.7% the etiology of the epilepsy was unknown. RESULTS Six months after initiating the ketogenic diet, it was effective (greater than a 50% reduction in seizure frequency) in 31.6% of patients. We did not find a link between rate of efficacy for the ketogenic diet and etiologies of epilepsy, nor did we find a link between the rate of efficacy for the ketogenic diet and cerebrospinal fluid pterins and biogenic amines concentrations. However, we found statistically significant differences for lysine and arginine values in the cerebrospinal fluid between ketogenic diet responders and nonresponders, but not for the other amino acids analyzed. SIGNIFICANCE The values of some amino acids were significantly different in relationship with the ketogenic diet efficacy; however, the epilepsy etiology and the cerebrospinal fluid biogenic amine and pterin values were not.
Collapse
|
36
|
Ferey L, Delaunay N. Food Analysis on Electrophoretic Microchips. SEPARATION AND PURIFICATION REVIEWS 2015. [DOI: 10.1080/15422119.2015.1014049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Karamikhah R, Jamshidzadeh A, Azarpira N, Saeidi A, Heidari R. Propylthiouracil-Induced Liver Injury in Mice and the Protective Role of Taurine. PHARMACEUTICAL SCIENCES 2015. [DOI: 10.15171/ps.2015.23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
38
|
Konstantakopoulos G, Dimitrakopoulos S, Michalopoulou PG. Drugs under early investigation for the treatment of bipolar disorder. Expert Opin Investig Drugs 2015; 24:477-90. [DOI: 10.1517/13543784.2015.1019061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- George Konstantakopoulos
- 1University of Athens, Eginition Hospital, First Department of Psychiatry, 72-74 Vas. Sofias Avenue, 11528, Athens, Greece ;
- 2Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Department of Psychosis Studies, London, UK
| | - Stefanos Dimitrakopoulos
- 1University of Athens, Eginition Hospital, First Department of Psychiatry, 72-74 Vas. Sofias Avenue, 11528, Athens, Greece ;
| | - Panayiota G Michalopoulou
- 2Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Department of Psychosis Studies, London, UK
| |
Collapse
|
39
|
Heidari R, Jamshidzadeh A, Keshavarz N, Azarpira N. Mitigation of Methimazole-Induced Hepatic Injury by Taurine in Mice. Sci Pharm 2014; 83:143-58. [PMID: 26839807 PMCID: PMC4727863 DOI: 10.3797/scipharm.1408-04] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/30/2014] [Indexed: 12/18/2022] Open
Abstract
Methimazole is the most widely prescribed antithyroid medication in humans. However, hepatotoxicity is a deleterious adverse effect associated with methimazole administration. No specific protective agent has been developed against this complication yet. This study was designed to investigate the role of taurine as a hepatoprotective agent against methimazole-induced liver injury in mice. Different reactive metabolites were proposed to be responsible for methimazole hepatotoxicity. Hence, methimazole-induced liver injury was investigated in intact and/or enzyme-induced animals in the current investigation. Animals were treated with methimazole (200 mg/kg, by gavage), and hepatic injury induced by this drug was investigated in intact and/or enzyme-induced groups. Markers such as lipid peroxidation, hepatic glutathione content, alanine aminotransferase (ALT) and alkaline phosphatase (ALP) in plasma, and histopathological changes in the liver of animals were monitored after drug administration. Methimazole caused liver injury as revealed by increased plasma ALT. Furthermore, a significant amount of lipid peroxidation was detected in the drug-treated animals, and hepatic glutathione reservoirs were depleted. Methimazole-induced hepatotoxicity was more severe in enzyme-induced mice. The above-mentioned alterations in hepatotoxicity markers were endorsed by significant histopathological changes in the liver. Taurine administration (1 g/kg, i.p.) effectively alleviated methimazole-induced liver injury in both intact and/or enzyme-induced animals.
Collapse
Affiliation(s)
- Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 7146864685 Shiraz, Iran
| | - Akram Jamshidzadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 7146864685 Shiraz, Iran; Pharmacology and Toxicology Department, Shiraz University of Medical Sciences, 7146864685 Shiraz, Iran
| | - Nahid Keshavarz
- Pharmacology and Toxicology Department, Shiraz University of Medical Sciences, 7146864685 Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, 7146864685 Shiraz, Iran
| |
Collapse
|
40
|
Larsen JA, Owens TJ, Fascetti AJ. Nutritional management of idiopathic epilepsy in dogs. J Am Vet Med Assoc 2014; 245:504-8. [DOI: 10.2460/javma.245.5.504] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
41
|
Chen R, Okabe A, Sun H, Sharopov S, Hanganu-Opatz IL, Kolbaev SN, Fukuda A, Luhmann HJ, Kilb W. Activation of glycine receptors modulates spontaneous epileptiform activity in the immature rat hippocampus. J Physiol 2014; 592:2153-68. [PMID: 24665103 DOI: 10.1113/jphysiol.2014.271700] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
While the expression of glycine receptors in the immature hippocampus has been shown, no information about the role of glycine receptors in controlling the excitability in the immature CNS is available. Therefore, we examined the effect of glycinergic agonists and antagonists in the CA3 region of an intact corticohippocampal preparation of the immature (postnatal days 4-7) rat using field potential recordings. Bath application of 100 μM taurine or 10 μM glycine enhanced the occurrence of recurrent epileptiform activity induced by 20 μM 4-aminopyridine in low Mg(2+) solution. This proconvulsive effect was prevented by 3 μM strychnine or after incubation with the loop diuretic bumetanide (10 μM), suggesting that it required glycine receptors and an active NKCC1-dependent Cl(-) accumulation. Application of higher doses of taurine (≥ 1 mM) or glycine (100 μM) attenuated recurrent epileptiform discharges. The anticonvulsive effect of taurine was also observed in the presence of the GABAA receptor antagonist gabazine and was attenuated by strychnine, suggesting that it was partially mediated by glycine receptors. Bath application of the glycinergic antagonist strychnine (0.3 μM) induced epileptiform discharges. We conclude from these results that in the immature hippocampus, activation of glycine receptors can mediate both pro- and anticonvulsive effects, but that a persistent activation of glycine receptors is required to suppress epileptiform activity. In summary, our study elucidated the important role of glycine receptors in the control of neuronal excitability in the immature hippocampus.
Collapse
Affiliation(s)
- Rongqing Chen
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| | - Akihito Okabe
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany Department of Molecular Anatomy, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Haiyan Sun
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| | - Salim Sharopov
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany University Medical Center Hamburg Eppendorf, Developmental Neurophysiology, Falkenried 94, D-20251, Hamburg, Germany
| | - Sergei N Kolbaev
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55120, Mainz, Germany
| |
Collapse
|