1
|
Gao Y, Liu N, Chen J, Zheng P, Niu J, Tang S, Peng X, Wu J, Yu J, Ma L. Neuropharmacological insight into preventive intervention in posttraumatic epilepsy based on regulating glutamate homeostasis. CNS Neurosci Ther 2023; 29:2430-2444. [PMID: 37309302 PMCID: PMC10401093 DOI: 10.1111/cns.14294] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 05/15/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Posttraumatic epilepsy (PTE) is one of the most critical complications of traumatic brain injury (TBI), significantly increasing TBI patients' neuropsychiatric symptoms and mortality. The abnormal accumulation of glutamate caused by TBI and its secondary excitotoxicity are essential reasons for neural network reorganization and functional neural plasticity changes, contributing to the occurrence and development of PTE. Restoring glutamate balance in the early stage of TBI is expected to play a neuroprotective role and reduce the risk of PTE. AIMS To provide a neuropharmacological insight for drug development to prevent PTE based on regulating glutamate homeostasis. METHODS We discussed how TBI affects glutamate homeostasis and its relationship with PTE. Furthermore, we also summarized the research progress of molecular pathways for regulating glutamate homeostasis after TBI and pharmacological studies aim to prevent PTE by restoring glutamate balance. RESULTS TBI can lead to the accumulation of glutamate in the brain, which increases the risk of PTE. Targeting the molecular pathways affecting glutamate homeostasis helps restore normal glutamate levels and is neuroprotective. DISCUSSION Taking glutamate homeostasis regulation as a means for new drug development can avoid the side effects caused by direct inhibition of glutamate receptors, expecting to alleviate the diseases related to abnormal glutamate levels in the brain, such as PTE, Parkinson's disease, depression, and cognitive impairment. CONCLUSION It is a promising strategy to regulate glutamate homeostasis through pharmacological methods after TBI, thereby decreasing nerve injury and preventing PTE.
Collapse
Affiliation(s)
- Yuan Gao
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Ning Liu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Juan Chen
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Ping Zheng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianguo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Xiaodong Peng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jing Wu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianqiang Yu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Lin Ma
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| |
Collapse
|
2
|
Bartkiene E, Tolpeznikaite E, Klupsaite D, Starkute V, Bartkevics V, Skrastina A, Pavlenko R, Mockus E, Lele V, Batkeviciute G, Budrikyte A, Janulyte R, Jomantaite I, Kybartaite A, Knystautaite K, Valionyte A, Ruibys R, Rocha JM. Bio-Converted Spirulina for Nutraceutical Chewing Candy Formulations Rich in L-Glutamic and Gamma-Aminobutyric Acids. Microorganisms 2023; 11:microorganisms11020441. [PMID: 36838408 PMCID: PMC9959499 DOI: 10.3390/microorganisms11020441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
This study aimed at evaluating changes of microalgae Spirulina during its fermentation with Lactiplantibacillus plantarum No. 122 strain, and further at incorporating Spirulina bio-converted for nutraceuticals rich in L-glutamic (L-Glu) and gamma-aminobutyric acids (GABA) into sucrose-free chewing candy (gummy) preparations. Fermented Spirulina had higher b* (yellowness) coordinates than untreated (non-fermented), and fermentation duration (24 and 48 h) had a statistically significant effect on colour coordinates. The highest contents of L-glutamic and gamma-aminobutyric acids (4062 and 228.6 mg/kg, respectively) were found in 24 and 48 h-fermented Spirulina, respectively. Fermentation increased the content of saturated fatty acids and omega-3 in Spirulina, while monounsaturated fatty acids and omega-6 were reduced. The addition of fermented Spirulina (FSp) significantly affected hardness, decreased lightness and yellowness, and increased the greenness of chewing candies. All chewing candy samples (with xylitol) prepared with 3 and 5 g of FSp and 0.2 µL of Citrus paradise essential oil received the highest scores for overall acceptability, and the highest intensity (0.052) of emotion "happy" was elicited by the sample group containing xylitol, agar, ascorbic acid, 3 g of FSp, and 0.1 µL of Mentha spicata essential oil. As an outcome of this research, one may conclude that fermented Spirulina has significant potential as an innovative ingredient in the production of healthier sucrose-free nutraceutical chewing candies.
Collapse
Affiliation(s)
- Elena Bartkiene
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Correspondence: ; Tel.: +370-601-35837
| | - Ernesta Tolpeznikaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Dovile Klupsaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Vytaute Starkute
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Vadims Bartkevics
- Institute of Food Safety, Animal Health and Environment “BIOR”, Zemgales Priekšpilsēta, LV-1076 Riga, Latvia
| | - Anna Skrastina
- Institute of Food Safety, Animal Health and Environment “BIOR”, Zemgales Priekšpilsēta, LV-1076 Riga, Latvia
| | - Romans Pavlenko
- Institute of Food Safety, Animal Health and Environment “BIOR”, Zemgales Priekšpilsēta, LV-1076 Riga, Latvia
| | - Ernestas Mockus
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Vita Lele
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Gabija Batkeviciute
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Ausrine Budrikyte
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Rusne Janulyte
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Ieva Jomantaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Auguste Kybartaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Karolina Knystautaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Aiste Valionyte
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Romas Ruibys
- Institute of Agricultural and Food Sciences, Agriculture Academy, Vytautas Magnus University, 44307 Kaunas, Lithuania
| | - João Miguel Rocha
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Department of Chemical Engineering (DEQ), Faculty of Engineering, University of Porto (FEUP), Rua Roberto Frias, s/n, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| |
Collapse
|
3
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- 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 111711, Colombia
| |
Collapse
|
4
|
Kovács Z, Skatchkov SN, Szabó Z, Qahtan S, Méndez-González MP, Malpica-Nieves CJ, Eaton MJ, Kardos J, Héja L. Putrescine Intensifies Glu/GABA Exchange Mechanism and Promotes Early Termination of Seizures. Int J Mol Sci 2022; 23:ijms23158191. [PMID: 35897767 PMCID: PMC9331600 DOI: 10.3390/ijms23158191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Endogenous anticonvulsant mechanisms represent a reliable and currently underdeveloped strategy against recurrent seizures and may recall novel original therapeutics. Here, we investigated whether the intensification of the astroglial Glu-GABA exchange mechanism by application of the GABA precursor putrescine (PUT) may be effective against convulsive and non-convulsive seizures. We explored the potential of PUT to inhibit spontaneous spike-and-wave discharges (SWDs) in WAG/Rij rats, a genetic model of absence epilepsy. Significant shortening of SWDs in response to intraperitoneally applied PUT has been observed, which could be antagonized by blocking GAT-2/3-mediated astrocytic GABA release with the specific inhibitor SNAP-5114. Direct application of exogenous GABA also reduced SWD duration, suggesting that PUT-triggered astroglial GABA release through GAT-2/3 may be a critical step in limiting seizure duration. PUT application also dose-dependently shortened seizure-like events (SLEs) in the low-[Mg2+] in vitro model of temporal lobe epilepsy. SNAP-5114 reversed the antiepileptic effect of PUT in the in vitro model as well, further confirming that PUT reduces seizure duration by triggering glial GABA release. In accordance, we observed that PUT specifically reduces the frequency of excitatory synaptic potentials, suggesting that it specifically acts at excitatory synapses. We also identified that PUT specifically eliminated the tonic depolarization-induced desynchronization of SLEs. Since PUT is an important source of glial GABA and we previously showed significant GABA release, it is suggested that the astroglial Glu-GABA exchange mechanism plays a key role in limiting ictal discharges, potentially opening up novel pathways to control seizure propagation and generalization.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Biology, Savaria University Centre, ELTE Eötvös Loránd University, Károlyi Gáspár tér 4, 9700 Szombathely, Hungary;
| | - Serguei N. Skatchkov
- Department of Physiology, Universidad Central del Caribe, Bayamon, PR 00960, USA; (S.N.S.); (C.J.M.-N.)
- Department of Biochemistry, Universidad Central del Caribe, Bayamon, PR 00960, USA; (M.P.M.-G.); (M.J.E.)
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary; (Z.S.); (S.Q.); (J.K.)
| | - Saif Qahtan
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary; (Z.S.); (S.Q.); (J.K.)
- Hevesy György PhD School of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117 Budapest, Hungary
- College of Science, University of Al-Qadisiyah, Al-Diwaniyah 58001, Iraq
| | - Miguel P. Méndez-González
- Department of Biochemistry, Universidad Central del Caribe, Bayamon, PR 00960, USA; (M.P.M.-G.); (M.J.E.)
- Natural Sciences Department, University of Puerto Rico in Aguadilla, Aguadilla, PR 00604, USA
- Department of Science and Technology, Antilles Adventist University, Mayagüez, PR 00681, USA
| | - Christian J. Malpica-Nieves
- Department of Physiology, Universidad Central del Caribe, Bayamon, PR 00960, USA; (S.N.S.); (C.J.M.-N.)
- Department of Biochemistry, Universidad Central del Caribe, Bayamon, PR 00960, USA; (M.P.M.-G.); (M.J.E.)
| | - Misty J. Eaton
- Department of Biochemistry, Universidad Central del Caribe, Bayamon, PR 00960, USA; (M.P.M.-G.); (M.J.E.)
| | - Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary; (Z.S.); (S.Q.); (J.K.)
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary; (Z.S.); (S.Q.); (J.K.)
- Correspondence:
| |
Collapse
|
5
|
Uddin MS, Lim LW. Glial cells in Alzheimer's disease: From neuropathological changes to therapeutic implications. Ageing Res Rev 2022; 78:101622. [PMID: 35427810 DOI: 10.1016/j.arr.2022.101622] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that usually develops slowly and progressively worsens over time. Although there has been increasing research interest in AD, its pathogenesis is still not well understood. Although most studies primarily focus on neurons, recent research findings suggest that glial cells (especially microglia and astrocytes) are associated with AD pathogenesis and might provide various possible therapeutic targets. Growing evidence suggests that microglia can provide protection against AD pathogenesis, as microglia with weakened functions and impaired responses to Aβ proteins are linked with elevated AD risk. Interestingly, numerous findings also suggest that microglial activation can be detrimental to neurons. Indeed, microglia can induce synapse loss via the engulfment of synapses, possibly through a complement-dependent process. Furthermore, they can worsen tau pathology and release inflammatory factors that cause neuronal damage directly or through the activation of neurotoxic astrocytes. Astrocytes play a significant role in various cerebral activities. Their impairment can mediate neurodegeneration and ultimately the retraction of synapses, resulting in AD-related cognitive deficits. Deposition of Aβ can result in astrocyte reactivity, which can further lead to neurotoxic effects and elevated secretion of inflammatory mediators and cytokines. Moreover, glial-induced inflammation in AD can exert both beneficial and harmful effects. Understanding the activities of astrocytes and microglia in the regulation of AD pathogenesis would facilitate the development of novel therapies. In this article, we address the implications of microglia and astrocytes in AD pathogenesis. We also discuss the mechanisms of therapeutic agents that exhibit anti-inflammatory effects against AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
6
|
Alavi MS, Fanoudi S, Hosseini M, Sadeghnia HR. Beneficial effects of levetiracetam in streptozotocin-induced rat model of Alzheimer's disease. Metab Brain Dis 2022; 37:689-700. [PMID: 35098412 DOI: 10.1007/s11011-021-00888-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder among the elderly. In the light of increasing AD prevalence and lack of effective treatment, new strategies to prevent or reverse this condition are needed. Levetiracetam (LEV) is a newer antiepileptic drug that is commonly used to treat certain types of seizures. Researches indicated that LEV has several other pharmacological activities, including improvement of cognitive function. In this study, the recovery effects of chronic (28 days) administration of LEV (50, 100, and 150 mg/kg, ip) on cognitive deficits caused by the intracerebroventricular (icv) injection of streptozotocin (STZ), as a model for sporadic AD, were evaluated in rats. We also considered the protective effects of LEV against hippocampal cell loss, oxidative damage, acetylcholinesterase (AChE) activity, neuroinflammation, and tauopathy caused by STZ. LEV (100 and 150 mg/kg) significantly attenuated the STZ-induced learning and memory impairments in the passive avoidance and Morris water maze (MWM) tasks. In addition, LEV suppressed STZ-induced hippocampal neuronal loss, while restored alterations in the redox status (lipid peroxides and glutathione), AChE activity, proinflammatory cytokines (IL-1β, IL-6, TNF-α), and hyperphosphorylation of tau linked to STZ administration. In conclusion, our study demonstrated that LEV alleviated hippocampal cell death and memory deficits in STZ-AD rats, through mitigating oxidative damage, suppression of proinflammatory cytokines expression, and inhibition of abnormal tau hyperphosphorylation.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Fanoudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid R Sadeghnia
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Kovács Z, Skatchkov SN, Veh RW, Szabó Z, Németh K, Szabó PT, Kardos J, Héja L. Critical Role of Astrocytic Polyamine and GABA Metabolism in Epileptogenesis. Front Cell Neurosci 2022; 15:787319. [PMID: 35069115 PMCID: PMC8770812 DOI: 10.3389/fncel.2021.787319] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence indicate that astrocytes are essential players of the excitatory and inhibitory signaling during normal and epileptiform activity via uptake and release of gliotransmitters, ions, and other substances. Polyamines can be regarded as gliotransmitters since they are almost exclusively stored in astrocytes and can be released by various mechanisms. The polyamine putrescine (PUT) is utilized to synthesize GABA, which can also be released from astrocytes and provide tonic inhibition on neurons. The polyamine spermine (SPM), synthesized form PUT through spermidine (SPD), is known to unblock astrocytic Cx43 gap junction channels and therefore facilitate astrocytic synchronization. In addition, SPM released from astrocytes may also modulate neuronal NMDA, AMPA, and kainate receptors. As a consequence, astrocytic polyamines possess the capability to significantly modulate epileptiform activity. In this study, we investigated different steps in polyamine metabolism and coupled GABA release to assess their potential to control seizure generation and maintenance in two different epilepsy models: the low-[Mg2+] model of temporal lobe epilepsy in vitro and in the WAG/Rij rat model of absence epilepsy in vivo. We show that SPM is a gliotransmitter that is released from astrocytes and significantly contributes to network excitation. Importantly, we found that inhibition of SPD synthesis completely prevented seizure generation in WAG/Rij rats. We hypothesize that this antiepileptic effect is attributed to the subsequent enhancement of PUT to GABA conversion in astrocytes, leading to GABA release through GAT-2/3 transporters. This interpretation is supported by the observation that antiepileptic potential of the Food and Drug Administration (FDA)-approved drug levetiracetam can be diminished by specifically blocking astrocytic GAT-2/3 with SNAP-5114, suggesting that levetiracetam exerts its effect by increasing surface expression of GAT-2/3. Our findings conclusively suggest that the major pathway through which astrocytic polyamines contribute to epileptiform activity is the production of GABA. Modulation of astrocytic polyamine levels, therefore, may serve for a more effective antiepileptic drug development in the future.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Biology, ELTE Eötvös Loránd University, Savaria University Centre, Szombathely, Hungary
| | - Serguei N. Skatchkov
- Department of Physiology, Universidad Central Del Caribe, Bayamon, PR, United States
- Department of Biochemistry, Universidad Central Del Caribe, Bayamon, PR, United States
| | - Rüdiger W. Veh
- Institut für Zell- und Neurobiologie, Centrum 2, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Krisztina Németh
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Pál T. Szabó
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| |
Collapse
|
8
|
Sharma P, Singh N, Singh S, Khare SK, Nain PKS, Nain L. Potent γ-amino butyric acid producing psychobiotic Lactococcus lactis LP-68 from non-rhizospheric soil of Syzygium cumini (Black plum). Arch Microbiol 2021; 204:82. [PMID: 34958412 DOI: 10.1007/s00203-021-02629-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/24/2022]
Abstract
Gamma amino butyric acid (GABA) is a chemical messenger that plays a significant role in muscle relaxation and brain health. Certain lactic acid bacteria (LAB) produce significant levels of GABA and thus act as potential psychobiotic cultures. In the present study, LAB were isolated from non-rhizospheric soil sample of Syzygium cumini (Black plum). A total of 57 LAB were isolated on the basis of their morphological and acid producing characteristic on de Man Rogosa Sharpe (MRS) agar. Only seven isolates were found to produce GABA (0.09-1.13 gL-1) in MRS broth and were identified as Lactococcus. However, L. lactis LP-68 produced highest amount of GABA and was selected for further optimization of culture conditions (pH, temperature and MSG) by response surface methodology (RSM). The optimization resulted in approximately four-fold increase in GABA production (4.11 gL-1). The results indicate that the L. lactis LP-68 can be used as starter culture for production of GABA-enriched functional foods.
Collapse
Affiliation(s)
- Pushpendra Sharma
- Division of Microbiology, ICAR-Indian Agriculture Research Institute, New Delhi, 110012, India
| | - Neera Singh
- Division of Agricultural Chemicals, ICAR-Indian Agriculture Research Institute, New Delhi, India
| | - Surender Singh
- Department of Microbiology, Central University of Haryana, Mahendergarh, 123031, India
| | - Sunil Kumar Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Pawan Kumar Singh Nain
- Design and Mechatronic Division, School of Civil and Mechanical Engineering, Galgotias University, Greater Noida, Gautam Budh Nagar, Uttar Pradesh, 201310, India
| | - Lata Nain
- Division of Microbiology, ICAR-Indian Agriculture Research Institute, New Delhi, 110012, India.
| |
Collapse
|
9
|
An Overview of Bioprocesses Employing Specifically Selected Microbial Catalysts for γ-Aminobutyric Acid Production. Microorganisms 2021; 9:microorganisms9122457. [PMID: 34946060 PMCID: PMC8704203 DOI: 10.3390/microorganisms9122457] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is an important chemical compound in the human brain. GABA acts as an inhibitory neurotransmitter by inducing hyperpolarization of cellular membranes. Usually, this pharmaceutically important compound is synthesized using a chemical process, but in this short overview we have only analysed microbial processes, which have been studied for the biosynthesis of this commercially important compound. The content of this article includes the following summarised information: the search for biological processes showed a number of lactic acid bacteria and certain species of fungi, which could be effectively used for the production of GABA. Strains found to possess GABA-producing pathways include Lactobacillus brevis CRL 1942, L. plantarum FNCC 260, Streptococcus salivarius subsp. thermophilus Y2, Bifidobacterium strains, Monascus spp., and Rhizopus spp. Each of these strains required specific growth conditions. However, several factors were common among these strains, such as the use of two main supplements in their fermentation medium—monosodium glutamate and pyridoxal phosphate—and maintaining an acidic pH. Optimization studies of GABA production were comprised of altering the media constituents, modifying growth conditions, types of cultivation system, and genetic manipulation. Some strains increased the production of GABA under anaerobic conditions. Genetic manipulation focused on silencing some genes or overexpression of gadB and gadC. The conclusion, based on the review of information available in published research, is that the targeted manipulation of selected microorganisms, as well as the culture conditions for an optimised bioprocess, should be adopted for an increased production of GABA to meet its increasing demand for food and pharmaceutical applications.
Collapse
|
10
|
Lin BC, Johal J, Sivakumar K, Romano AE, Yacoub HA. Stiff-person syndrome: an atypical presentation and a review of the literature. Hosp Pract (1995) 2021; 49:384-390. [PMID: 34313523 DOI: 10.1080/21548331.2021.1961456] [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] [Indexed: 10/20/2022]
Abstract
Introduction: Stiff-person syndrome (SPS) is a rare autoimmune neurological disorder associated with muscle rigidity and spasms. A number of antibodies have been associated with disorder, including anti-glutamic acid decarboxylase and anti-amphiphysin.Case report; In this report, we present a rare case of a 79-year-old woman who presented with bilateral lower extremity weakness who was ultimately diagnosed with stiff-limb syndrome, a rare variant of SPS. Extensive laboratory and CSF studies were unrevealing. Electromyography showed significant peroneal motor neuropathy and complex repetitive discharges in the left tibialis anterior muscle. Antibodies to glutamic acid decarboxylase were significantly elevated at 124 units/mL. She was subsequently started on oral diazepam with significant improvement in her symptoms.Conclusion: The presentation of SPS can vary based on epidemiologic factors, clinical symptoms, and associated disorders. These forms can have overlapping features which may make the categorization of patients into one of these forms challenging.
Collapse
Affiliation(s)
- Benjamin C Lin
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jaspreet Johal
- Department of Neurology, Lehigh Valley Health Network, Allentown, PA, USA
| | - Keithan Sivakumar
- Division of Neurology, Sunnybrook Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alissa E Romano
- Department of Neurology, Lehigh Valley Health Network, Allentown, PA, USA.,Department of Neurology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hussam A Yacoub
- Department of Neurology, Lehigh Valley Health Network, Allentown, PA, USA.,Department of Neurology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
11
|
Asanuma M, Miyazaki I. Glutathione and Related Molecules in Parkinsonism. Int J Mol Sci 2021; 22:ijms22168689. [PMID: 34445395 PMCID: PMC8395390 DOI: 10.3390/ijms22168689] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis, and release in/from surrounding astrocytes. Transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), a detoxifying master transcription factor, is expressed mainly in astrocytes and activates the gene expression of various phase II drug-metabolizing enzymes or antioxidants including GSH-related molecules and metallothionein by binding to the antioxidant response element (ARE) of these genes. Accumulating evidence has shown the involvement of dysfunction of antioxidative molecules including GSH and its related molecules in the pathogenesis of Parkinson’s disease (PD) or parkinsonian models. Furthermore, we found several agents targeting GSH synthesis in the astrocytes that protect nigrostriatal dopaminergic neuronal loss in PD models. In this article, the neuroprotective effects of supplementation and enhancement of GSH and its related molecules in PD pathology are reviewed, along with introducing new experimental findings, especially targeting of the xCT-GSH synthetic system and Nrf2–ARE pathway in astrocytes.
Collapse
|
12
|
Lange F, Hörnschemeyer J, Kirschstein T. Glutamatergic Mechanisms in Glioblastoma and Tumor-Associated Epilepsy. Cells 2021; 10:cells10051226. [PMID: 34067762 PMCID: PMC8156732 DOI: 10.3390/cells10051226] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022] Open
Abstract
The progression of glioblastomas is associated with a variety of neurological impairments, such as tumor-related epileptic seizures. Seizures are not only a common comorbidity of glioblastoma but often an initial clinical symptom of this cancer entity. Both, glioblastoma and tumor-associated epilepsy are closely linked to one another through several pathophysiological mechanisms, with the neurotransmitter glutamate playing a key role. Glutamate interacts with its ionotropic and metabotropic receptors to promote both tumor progression and excitotoxicity. In this review, based on its physiological functions, our current understanding of glutamate receptors and glutamatergic signaling will be discussed in detail. Furthermore, preclinical models to study glutamatergic interactions between glioma cells and the tumor-surrounding microenvironment will be presented. Finally, current studies addressing glutamate receptors in glioma and tumor-related epilepsy will be highlighted and future approaches to interfere with the glutamatergic network are discussed.
Collapse
Affiliation(s)
- Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany;
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
- Correspondence: (F.L.); (T.K.)
| | - Julia Hörnschemeyer
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany;
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
- Correspondence: (F.L.); (T.K.)
| |
Collapse
|
13
|
Levetiracetam treatment leads to functional recovery after thoracic or cervical injuries of the spinal cord. NPJ Regen Med 2021; 6:11. [PMID: 33654068 PMCID: PMC7977146 DOI: 10.1038/s41536-021-00121-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Spinal cord injury (SCI) leads to dramatic impairments of motor, sensory, and autonomic functions of affected individuals. Following the primary injury, there is an increased release of glutamate that leads to excitotoxicity and further neuronal death. Therefore, modulating glutamate excitotoxicity seems to be a promising target to promote neuroprotection during the acute phase of the injury. In this study, we evaluated the therapeutic effect of a FDA approved antiepileptic drug (levetiracetam-LEV), known for binding to the synaptic vesicle protein SV2A in the brain and spinal cord. LEV therapy was tested in two models of SCI-one affecting the cervical and other the thoracic level of the spinal cord. The treatment was effective on both SCI models. Treated animals presented significant improvements on gross and fine motor functions. The histological assessment revealed a significant decrease of cavity size, as well as higher neuronal and oligodendrocyte survival on treated animals. Molecular analysis revealed that LEV acts by stabilizing the astrocytes allowing an effective uptake of the excess glutamate from the extracellular space. Overall, our results demonstrate that Levetiracetam may be a promising drug for acute management of SCI.
Collapse
|
14
|
Asanuma M, Miyazaki I. [Anti-oxidants in astrocytes as target of neuroprotection for Parkinson's disease]. Nihon Yakurigaku Zasshi 2021; 156:14-20. [PMID: 33390474 DOI: 10.1254/fpj.20071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Recently, it has been reported that dysfunction of astrocytes is involved vulnerability of neuronal cells in several neurological disorders. Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine is readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis and release in/from surrounding astrocytes. The expression and release of the zinc-binding protein metallothionein (MT) in astrocytes, which is a strong antioxidant, is induced and exerts neuroprotective in the case of dopaminergic neuronal damage. In addition, the transcription factor Nrf2 induces expression of MT-1 and GSH related molecules. We previously revealed that several antiepileptic drugs, serotonin 5-HT1A receptor agonists, plant-derived chemicals (phytochemicals) increased xCT expression, Nrf2 activation, GSH or MT expression and release in/from astrocytes, and exerted a neuroprotective effect against dopaminergic neurodegeneration in Parkinson's disease model. Our serial studies on neuroprotection via antioxidant defense mechanism of astrocytes have found three target molecular systems of astrocytes for neuroprotection: (1) xCT-GSH synthetic system, (2) Nrf2 system and (3) 5-HT1A receptor-Nrf2-MT system, 5-HT1A-S100β system. In this article, possible neuroprotective strategy for Parkinson's disease has been reviewed targeting antioxidative molecules in astrocytes.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| |
Collapse
|
15
|
Deshpande LS, Blair RE, Halquist M, Kosmider L, DeLorenzo RJ. Intramuscular atenolol and levetiracetam reduce mortality in a rat model of paraoxon-induced status epilepticus. Ann N Y Acad Sci 2020; 1480:219-232. [PMID: 32961584 DOI: 10.1111/nyas.14500] [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: 04/24/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/01/2022]
Abstract
Organophosphorus (OP) compounds are chemical threat agents and are irreversible inhibitors of the enzyme acetylcholinesterase that lead to a hypercholinergic response that could include status epilepticus (SE). SE particularly targets the heart and brain and despite existing therapies, it is still associated with significant mortality and morbidity. Here, we investigated the effect of intramuscular (i.m.) adjunct therapy consisting of atenolol (AT) and levetiracetam (LV) when administered after paraoxon (POX)-induced SE. The combination therapy was administered twice daily for 2, 7, or 14 days. POX exposure in rats produced rapid SE onset that was treated with atropine, pralidoxime chloride, and midazolam. Here, AT + LV therapy produced significant reductions in POX SE mortality assessed at 30 days post-SE. AT + LV therapy exhibited muscle pathology inflammation scores that were not significantly different from saline-treated controls. Pharmacokinetic analyses revealed that the i.m. route achieved faster and stabler plasma therapeutic levels for both AT and LV under OP SE conditions compared with oral administrations. Our data provide evidence of the safety and efficacy of i.m. AT + LV therapy for reducing mortality following POX SE.
Collapse
Affiliation(s)
- Laxmikant S Deshpande
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia.,Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Robert E Blair
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Matthew Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Leon Kosmider
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Robert J DeLorenzo
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia.,Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
16
|
Klein P, Friedman A, Hameed MQ, Kaminski RM, Bar-Klein G, Klitgaard H, Koepp M, Jozwiak S, Prince DA, Rotenberg A, Twyman R, Vezzani A, Wong M, Löscher W. Repurposed molecules for antiepileptogenesis: Missing an opportunity to prevent epilepsy? Epilepsia 2020; 61:359-386. [PMID: 32196665 PMCID: PMC8317585 DOI: 10.1111/epi.16450] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Prevention of epilepsy is a great unmet need. Acute central nervous system (CNS) insults such as traumatic brain injury (TBI), cerebrovascular accidents (CVA), and CNS infections account for 15%-20% of all epilepsy. Following TBI and CVA, there is a latency of days to years before epilepsy develops. This allows treatment to prevent or modify postinjury epilepsy. No such treatment exists. In animal models of acquired epilepsy, a number of medications in clinical use for diverse indications have been shown to have antiepileptogenic or disease-modifying effects, including medications with excellent side effect profiles. These include atorvastatin, ceftriaxone, losartan, isoflurane, N-acetylcysteine, and the antiseizure medications levetiracetam, brivaracetam, topiramate, gabapentin, pregabalin, vigabatrin, and eslicarbazepine acetate. In addition, there are preclinical antiepileptogenic data for anakinra, rapamycin, fingolimod, and erythropoietin, although these medications have potential for more serious side effects. However, except for vigabatrin, there have been almost no translation studies to prevent or modify epilepsy using these potentially "repurposable" medications. We may be missing an opportunity to develop preventive treatment for epilepsy by not evaluating these medications clinically. One reason for the lack of translation studies is that the preclinical data for most of these medications are disparate in terms of types of injury, models within different injury type, dosing, injury-treatment initiation latencies, treatment duration, and epilepsy outcome evaluation mode and duration. This makes it difficult to compare the relative strength of antiepileptogenic evidence across the molecules, and difficult to determine which drug(s) would be the best to evaluate clinically. Furthermore, most preclinical antiepileptogenic studies lack information needed for translation, such as dose-blood level relationship, brain target engagement, and dose-response, and many use treatment parameters that cannot be applied clinically, for example, treatment initiation before or at the time of injury and dosing higher than tolerated human equivalent dosing. Here, we review animal and human antiepileptogenic evidence for these medications. We highlight the gaps in our knowledge for each molecule that need to be filled in order to consider clinical translation, and we suggest a platform of preclinical antiepileptogenesis evaluation of potentially repurposable molecules or their combinations going forward.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland
| | - Alon Friedman
- Departments of Physiology and Cell Biology, and Brain and Cognitive Science, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Departments of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Canada
| | - Mustafa Q. Hameed
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rafal M. Kaminski
- Neurosymptomatic Domains Section, Roche Pharma Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Guy Bar-Klein
- McKusick-Nathans Institute of Genetic Medicine, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henrik Klitgaard
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l’Alleud, Belgium
| | - Mathias Koepp
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Sergiusz Jozwiak
- Department of Pediatric Neurology, Warsaw Medical University, Warsaw, Poland
| | - David A. Prince
- Neurology and the Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Scientific Institute for Research and Health Care, Milan, Italy
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
17
|
Fattorini G, Melone M, Conti F. A Reappraisal of GAT-1 Localization in Neocortex. Front Cell Neurosci 2020; 14:9. [PMID: 32116556 PMCID: PMC7031676 DOI: 10.3389/fncel.2020.00009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
γ-Aminobutyric acid (GABA) transporter (GAT)-1, the major GABA transporter in the brain, plays a key role in modulating GABA signaling and is involved in the pathophysiology of several neuropsychiatric diseases, including epilepsy. The original description of GAT-1 as a neuronal transporter has guided the interpretation of the findings of all physiological, pharmacological, genetic, or clinical studies. However, evidence published in the past few years, some of which is briefly reviewed herein, does not seem to be consistent with a neurocentric view of GAT-1 function and calls for more detailed analysis of its localization. We therefore performed a thorough systematic assessment of GAT-1 localization in neocortex and subcortical white matter. In line with earlier work, we found that GAT-1 was robustly expressed in axon terminals forming symmetric synapses and in astrocytic processes, whereas its astrocytic expression was more diffuse than expected and, even more surprisingly, immature and mature oligodendrocytes and microglial cells also expressed the transporter. These data indicate that the era of “neuronal” and “glial” GABA transporters has finally come to a close and provide a wider perspective from which to view GABA-mediated physiological phenomena. In addition, given the well-known involvement of astrocytes, oligodendrocytes, and microglial cells in physiological as well as pathological conditions, the demonstration of functional GAT-1 in these cells is expected to provide greater insight into the phenomena occurring in the diseased brain as well as to prompt a reassessment of earlier findings.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.,Fondazione di Medicina Molecolare, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
18
|
Sadleir LG, Kolc KL, King C, Mefford HC, Dale RC, Gecz J, Scheffer IE. Levetiracetam efficacy in PCDH19 Girls Clustering Epilepsy. Eur J Paediatr Neurol 2020; 24:142-147. [PMID: 31928905 DOI: 10.1016/j.ejpn.2019.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 12/18/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND PCDH19 Girls clustering epilepsy (GCE) has a phenotypic spectrum that includes developmental and epileptic encephalopathy. PCDH19-GCE presents with clusters of seizures in the first years of life. Although patients typically outgrow their seizures, many are left with intellectual disability. Here we retrospectively assess the effect of levetiracetam in two independent cohorts of females with PCDH19-GCE. METHODS Cohort A was identified by searching our epilepsy genetics research database for girls with PCDH19-GCE who had trialled levetiracetam. Cohort B consisted of girls aged 2 years or older, including women, participating in an international online questionnaire. Information regarding seizure frequency and levetiracetam use was obtained by in-person patient interview and review of clinical records for cohort A, and by patient report for cohort B. RESULTS Cohort A consisted of 17 females, aged 3-37 years, who had a trial of levetiracetam at an average age of 10.7 years. 13/17 females became seizure free for >12 months; while 10/17 remained seizure free for >24 months. Cohort B comprised 62 females, aged 1.5-41 years. 26/62 became seizure free for >12 months, and 19/62 for >24 months on levetiracetam therapy. DISCUSSION Levetiracetam was effective in two cohorts of females with PCDH19-GCE where 42% and 76% of females became seizure free for >12 months, respectively. Levetiracetam is an effective therapy for females with PCDH19-GCE and should be considered early in the management of the highly refractory clusters of seizures that characterise this genetic disease.
Collapse
Affiliation(s)
- Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand.
| | - Kristy L Kolc
- Adelaide Medical School, The University of Adelaide, SA, Australia
| | - Chontelle King
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Heather C Mefford
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Russell C Dale
- Kids Neuroscience Centre. Children's Hospital at Westmead, University of Sydney, Australia
| | - Jozef Gecz
- Adelaide Medical School, The University of Adelaide, SA, Australia; Robinson Research Institute, The University of Adelaide, SA, Australia; Healthy Mothers and Babies, South Australian Health and Medical Research Institute, SA, Australia
| | - Ingrid E Scheffer
- Department of Medicine, Epilepsy Research Centre, The University of Melbourne, Austin Health, Melbourne, VIC, Australia; Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, VIC, Australia; The Florey Institute and Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
19
|
A Brief Review on the Non-protein Amino Acid, Gamma-amino Butyric Acid (GABA): Its Production and Role in Microbes. Curr Microbiol 2019; 77:534-544. [PMID: 31844936 DOI: 10.1007/s00284-019-01839-w] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
Gamma-Aminobutyric acid (GABA) is a non-protein amino acid widely distributed in nature. It is produced through irreversible α-decarboxylation of glutamate by enzyme glutamate decarboxylase (GAD). GABA and GAD have been found in plants, animals, and microorganisms. GABA is distributed throughout the human body and it is involved in the regulation of cardiovascular conditions such as blood pressure and heart rate, and plays a role in the reduction of anxiety and pain. Although researchers had produced GABA by chemical method earlier it became less acceptable as it pollutes the environment. Researchers now use a more promising microbial method for the production of GABA. In the drug and food industry, demand for GABA is immense. So, large scale conversion of GABA by microbes has got much attention. So this review focuses on the isolation source, production, and functions of GABA in the microbial system. We also summarize the mechanism of action of GABA and its shunt pathway.
Collapse
|
20
|
Héja L, Simon Á, Szabó Z, Kardos J. Feedback adaptation of synaptic excitability via Glu:Na + symport driven astrocytic GABA and Gln release. Neuropharmacology 2019; 161:107629. [PMID: 31103619 DOI: 10.1016/j.neuropharm.2019.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/30/2019] [Accepted: 05/07/2019] [Indexed: 02/08/2023]
Abstract
Glutamatergic transmission composed of the arriving of action potential at the axon terminal, fast vesicular Glu release, postsynaptic Glu receptor activation, astrocytic Glu clearance and Glu→Gln shuttle is an abundantly investigated phenomenon. Despite its essential role, however, much less is known about the consequences of the mechanistic connotations of Glu:Na+ symport. Due to the coupled Na+ transport, Glu uptake results in significantly elevated intracellular astrocytic [Na+] that markedly alters the driving force of other Na+-coupled astrocytic transporters. The resulting GABA and Gln release by reverse transport through the respective GAT-3 and SNAT3 transporters help to re-establish the physiological Na+ homeostasis without ATP dissipation and consequently leads to enhanced tonic inhibition and replenishment of axonal glutamate pool. Here, we place this emerging astrocytic adjustment of synaptic excitability into the centre of future perspectives. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary.
| |
Collapse
|
21
|
Mechanisms Underlying Aggressive Behavior Induced by Antiepileptic Drugs: Focus on Topiramate, Levetiracetam, and Perampanel. Behav Neurol 2018; 2018:2064027. [PMID: 30581496 PMCID: PMC6276511 DOI: 10.1155/2018/2064027] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/30/2018] [Indexed: 12/28/2022] Open
Abstract
Antiepileptic drugs (AEDs) are effective against seizures, but their use is often limited by adverse effects, among them psychiatric and behavioral ones including aggressive behavior (AB). Knowledge of the incidence, risk factors, and the underlying mechanisms of AB induced by AEDs may help to facilitate management and reduce the risk of such side effects. The exact incidence of AB as an adverse effect of AEDs is difficult to estimate, but frequencies up to 16% have been reported. Primarily, levetiracetam (LEV), perampanel (PER), and topiramate (TPM), which have diverse mechanisms of action, have been associated with AB. Currently, there is no evidence for a specific pharmacological mechanism solely explaining the increased incidence of AB with LEV, PER, and TPM. Serotonin (5-HT) and GABA, and particularly glutamate (via the AMPA receptor), seem to play key roles. Other mechanisms involve hormones, epigenetics, and “alternative psychosis” and related phenomena. Increased individual susceptibility due to an underlying neurological and/or a mental health disorder may further explain why people with epilepsy are at an increased risk of AB when using AEDs. Remarkably, AB may occur with a delay of weeks or months after start of treatment. Information to patients, relatives, and caregivers, as well as sufficient clinical follow-up, is crucial, and there is a need for further research to understand the complex relationship between AED mechanisms of action and the induction/worsening of AB.
Collapse
|
22
|
Pastukhov A, Borisova T. Levetiracetam-mediated improvement of decreased NMDA-induced glutamate release from nerve terminals during hypothermia. Brain Res 2018; 1699:69-78. [PMID: 30343685 DOI: 10.1016/j.brainres.2018.06.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/09/2018] [Accepted: 06/28/2018] [Indexed: 01/12/2023]
Abstract
A combination of a beneficial neuroprotectant, hypothermia, with targeted medication is a perspective therapeutic approach. Here, we analyzed both non-specific (deep and profound hypothermia, 27 °C and 17 °C, respectively) and targeted (anticonvulsant drug levetiracetam) modulation of l-[14C]glutamate release induced by activation of presynaptic NMDA, AMPA, and kainate receptors in rat brain nerve terminals (synaptosomes). Gradual dynamics of hypothermia-mediated decrease in synaptosomal l-[14C]glutamate release evoked by the receptor agonists NMDA-, AMPA-, and kainate (250 μM) has been demonstrated that can be of value for the justification of optimal temperature regimes in therapeutic hypothermia. 250 μM NMDA-induced l-[14C]glutamate release from nerve terminals was higher in the presence of levetiracetam (100 μM) as compared to that without the drug. Despite levetiracetam effects decreased in hypothermia, combined application of hypothermia and levetiracetam resulted in higher NMDA-induced l-[14C]glutamate release from nerve terminals as compared to that without the drug. These effects were not revealed for synaptosomal AMPA- and kainate-induced l-[14C]glutamate release in the presence of levetiracetam at the similar concentration. Therefore, levetiracetam administration significantly mitigated a hypothermia-induced decrease in NMDA receptor response at the presynaptic level and can be used for the targeted neurocorrection to reduce side effects of hypothermia in cardiac surgery. However, levetiracetam-mediated improvement of NMDA receptor response is not applicable in stroke, brain trauma and neonatal asphyxia therapies, where the main neuroprotective action of hypothermia is associated with prevention of damaging consequence of pre-existing acute glutamate exitotoxicity.
Collapse
Affiliation(s)
- A Pastukhov
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9 Leontovicha Street, Kiev 01030, Ukraine.
| | - T Borisova
- The Department of Neurochemistry, Palladin Institute of Biochemistry, NAS of Ukraine, 9 Leontovicha Street, Kiev 01030, Ukraine.
| |
Collapse
|
23
|
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: 3.2] [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
|
24
|
González-Reyes RE, Nava-Mesa MO, Vargas-Sánchez K, Ariza-Salamanca D, Mora-Muñoz L. Involvement of Astrocytes in Alzheimer's Disease from a Neuroinflammatory and Oxidative Stress Perspective. Front Mol Neurosci 2017; 10:427. [PMID: 29311817 PMCID: PMC5742194 DOI: 10.3389/fnmol.2017.00427] [Citation(s) in RCA: 339] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer disease (AD) is a frequent and devastating neurodegenerative disease in humans, but still no curative treatment has been developed. Although many explicative theories have been proposed, precise pathophysiological mechanisms are unknown. Due to the importance of astrocytes in brain homeostasis they have become interesting targets for the study of AD. Changes in astrocyte function have been observed in brains from individuals with AD, as well as in AD in vitro and in vivo animal models. The presence of amyloid beta (Aβ) has been shown to disrupt gliotransmission, neurotransmitter uptake, and alter calcium signaling in astrocytes. Furthermore, astrocytes express apolipoprotein E and are involved in the production, degradation and removal of Aβ. As well, changes in astrocytes that precede other pathological characteristics observed in AD, point to an early contribution of astroglia in this disease. Astrocytes participate in the inflammatory/immune responses of the central nervous system. The presence of Aβ activates different cell receptors and intracellular signaling pathways, mainly the advanced glycation end products receptor/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, responsible for the transcription of pro-inflammatory cytokines and chemokines in astrocytes. The release of these pro-inflammatory agents may induce cellular damage or even stimulate the production of Aβ in astrocytes. Additionally, Aβ induces the appearance of oxidative stress (OS) and production of reactive oxygen species and reactive nitrogen species in astrocytes, affecting among others, intracellular calcium levels, NADPH oxidase (NOX), NF-κB signaling, glutamate uptake (increasing the risk of excitotoxicity) and mitochondrial function. Excessive neuroinflammation and OS are observed in AD, and astrocytes seem to be involved in both. The Aβ/NF-κB interaction in astrocytes may play a central role in these inflammatory and OS changes present in AD. In this paper, we also discuss therapeutic measures highlighting the importance of astrocytes in AD pathology. Several new therapeutic approaches involving phenols (curcumin), phytoestrogens (genistein), neuroesteroids and other natural phytochemicals have been explored in astrocytes, obtaining some promising results regarding cognitive improvements and attenuation of neuroinflammation. Novel strategies comprising astrocytes and aimed to reduce OS in AD have also been proposed. These include estrogen receptor agonists (pelargonidin), Bambusae concretio Salicea, Monascin, and various antioxidatives such as resveratrol, tocotrienol, anthocyanins, and epicatechin, showing beneficial effects in AD models.
Collapse
Affiliation(s)
- Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Daniel Ariza-Salamanca
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Laura Mora-Muñoz
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
25
|
Marcotulli D, Fattorini G, Bragina L, Perugini J, Conti F. Levetiracetam Affects Differentially Presynaptic Proteins in Rat Cerebral Cortex. Front Cell Neurosci 2017; 11:389. [PMID: 29311825 PMCID: PMC5732259 DOI: 10.3389/fncel.2017.00389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/24/2017] [Indexed: 01/23/2023] Open
Abstract
Presynaptic proteins are potential therapeutic targets for epilepsy and other neurological diseases. We tested the hypothesis that chronic treatment with the SV2A ligand levetiracetam affects the expression of other presynaptic proteins. Results showed that in rat neocortex no significant difference was detected in SV2A protein levels in levetiracetam treated animals compared to controls, whereas levetiracetam post-transcriptionally decreased several vesicular proteins and increased LRRK2, without any change in mRNA levels. Analysis of SV2A interactome indicates that the presynaptic proteins regulation induced by levetiracetam reported here is mediated by this interactome, and suggests that LRRK2 plays a role in forging the pattern of effects.
Collapse
Affiliation(s)
- Daniele Marcotulli
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| | - Luca Bragina
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| |
Collapse
|
26
|
Vossel KA, Tartaglia MC, Nygaard HB, Zeman AZ, Miller BL. Epileptic activity in Alzheimer's disease: causes and clinical relevance. Lancet Neurol 2017; 16:311-322. [PMID: 28327340 DOI: 10.1016/s1474-4422(17)30044-3] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 01/18/2023]
Abstract
Epileptic activity is frequently associated with Alzheimer's disease; this association has therapeutic implications, because epileptic activity can occur at early disease stages and might contribute to pathogenesis. In clinical practice, seizures in patients with Alzheimer's disease can easily go unrecognised because they usually present as non-motor seizures, and can overlap with other symptoms of the disease. In patients with Alzheimer's disease, seizures can hasten cognitive decline, highlighting the clinical relevance of early recognition and treatment. Some evidence indicates that subclinical epileptiform activity in patients with Alzheimer's disease, detected by extended neurophysiological monitoring, can also lead to accelerated cognitive decline. Treatment of clinical seizures in patients with Alzheimer's disease with select antiepileptic drugs (AEDs), in low doses, is usually well tolerated and efficacious. Moreover, studies in mouse models of Alzheimer's disease suggest that certain classes of AEDs that reduce network hyperexcitability have disease-modifying properties. These AEDs target mechanisms of epileptogenesis involving amyloid β and tau. Clinical trials targeting network hyperexcitability in patients with Alzheimer's disease will identify whether AEDs or related strategies could improve their cognitive symptoms or slow decline.
Collapse
Affiliation(s)
- Keith A Vossel
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | | | - Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Adam Z Zeman
- Cognitive Neurology Research Group, University of Exeter Medical School, Exeter, UK
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
27
|
Sendrowski K, Sobaniec P, Poskrobko E, Rusak M, Sobaniec W. Unfavorable effect of levetiracetam on cultured hippocampal neurons after hyperthermic injury. Pharmacol Rep 2017; 69:462-468. [PMID: 31994103 DOI: 10.1016/j.pharep.2017.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND The aim of this study was to examine the viability of neurons and the putative neuroprotective effects of second-generation antiepileptic drug, levetiracetam (LEV), on cultured hippocampal neurons injured by hyperthermia. METHODS Primary cultures of rat's hippocampal neurons at 7 day in vitro (DIV) were incubated in the presence or absence of LEV in varied concentrations under hyperthermic conditions. Cultures were heated in a temperature of 40 °C for 24 h or in a temperature of 41 °C for 6 h. Flow cytometry with Annexin V/PI staining as well as fluorescent microscopy assay were used for counting and establishing neurons as viable, necrotic or apoptotic. Additionally, the release of lactate dehydrogenase (LDH) to the culture medium, as a marker of cell death, was evaluated. Assessment was performed after 9DIV and 10 DIV. RESULTS Incubation of hippocampal cultures in hyperthermic conditions resulted in statistically significant increase in the number of injured neurons when compared with non-heated control cultures. Intensity of neuronal destruction was dependent on temperature-value. When incubation temperature 40 °C was used, over 80% of the population of neurons remained viable after 10 DIV. Under higher temperature 41 °C, only less than 60% of neurons were viable after 10 DIV. Both apoptotic and necrotic pathways of neuronal death induced by hyperthermia were confirmed by Annexin V/PI staining. CONCLUSIONS LEV showed no neuroprotective effects in the current model of hyperthermia in vitro. Moreover, drug, especially when used in higher concentrations, exerted unfavorable intensification of aponecrosis of cultured hippocampal neurons.
Collapse
Affiliation(s)
- Krzysztof Sendrowski
- Department of Pediatric Neurology and Rehabilitation, Medical University of Bialystok, Białystok, Poland.
| | - Piotr Sobaniec
- Department of Pediatric Neurology and Rehabilitation, Medical University of Bialystok, Białystok, Poland
| | - Elżbieta Poskrobko
- Department of Pediatric Laboratory Diagnostic, Medical University of Bialystok, Białystok, Poland
| | - Małgorzata Rusak
- Department of Hematological Diagnostics, Medical University of Bialystok, Białystok, Poland
| | - Wojciech Sobaniec
- Department of Pediatric Neurology and Rehabilitation, Medical University of Bialystok, Białystok, Poland
| |
Collapse
|
28
|
Kardos J, Héja L, Jemnitz K, Kovács R, Palkovits M. The nature of early astroglial protection-Fast activation and signaling. Prog Neurobiol 2017; 153:86-99. [PMID: 28342942 DOI: 10.1016/j.pneurobio.2017.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/22/2016] [Accepted: 03/05/2017] [Indexed: 12/14/2022]
Abstract
Our present review is focusing on the uniqueness of balanced astroglial signaling. The balance of excitatory and inhibitory signaling within the CNS is mainly determined by sharp synaptic transients of excitatory glutamate (Glu) and inhibitory γ-aminobutyrate (GABA) acting on the sub-second timescale. Astroglia is involved in excitatory chemical transmission by taking up i) Glu through neurotransmitter-sodium transporters, ii) K+ released due to presynaptic action potential generation, and iii) water keeping osmotic pressure. Glu uptake-coupled Na+ influx may either ignite long-range astroglial Ca2+ transients or locally counteract over-excitation via astroglial GABA release and increased tonic inhibition. Imbalance of excitatory and inhibitory drives is associated with a number of disease conditions, including prevalent traumatic and ischaemic injuries or the emergence of epilepsy. Therefore, when addressing the potential of early therapeutic intervention, astroglial signaling functions combating progress of Glu excitotoxicity is of critical importance. We suggest, that excitotoxicity is linked primarily to over-excitation induced by the impairment of astroglial Glu uptake and/or GABA release. Within this framework, we discuss the acute alterations of Glu-cycling and metabolism and conjecture the therapeutic promise of regulation. We also confer the role played by key carrier proteins and enzymes as well as their interplay at the molecular, cellular, and organ levels. Moreover, based on our former studies, we offer potential prospect on the emerging theme of astroglial succinate sensing in course of Glu excitotoxicity.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary.
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary
| | - Richárd Kovács
- Institute of Neurophysiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| |
Collapse
|
29
|
Miyazaki I, Murakami S, Torigoe N, Kitamura Y, Asanuma M. Neuroprotective effects of levetiracetam target xCT in astrocytes in parkinsonian mice. J Neurochem 2015; 136:194-204. [DOI: 10.1111/jnc.13405] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Ikuko Miyazaki
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Shinki Murakami
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- SAIDO Co.; Fukuoka Japan
| | - Nao Torigoe
- Department of Clinical Pharmacy; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Yoshihisa Kitamura
- Department of Clinical Pharmacy; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | - Masato Asanuma
- Department of Brain Science; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
- Department of Medical Neurobiology; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| |
Collapse
|
30
|
Abstract
We delineate perspectives for the design and discovery of antiepileptic drugs (AEDs) with fewer side effects by focusing on astroglial modulation of spatiotemporal seizure dynamics. It is now recognized that the major inhibitory neurotransmitter of the brain, γ-aminobutyric acid (GABA), can be released through the reversal of astroglial GABA transporters. Synaptic spillover and subsequent glutamate (Glu) uptake in neighboring astrocytes evoke replacement of extracellular Glu for GABA, driving neurons away from the seizure threshold. Attenuation of synaptic signaling by this negative feedback through the interplay of Glu and GABA transporters of adjacent astroglia can result in shortened seizures. By contrast, long-range activation of astroglia through gap junctions may promote recurrent seizures on the model of pharmacoresistant temporal lobe epilepsy. From their first detection to our current understanding, we identify various targets that shape both short- and long-range neuro-astroglia coupling, as these are manifest in epilepsy phenomena and in the associated research promotions of AED.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117 Budapest, Hungary
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117 Budapest, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117 Budapest, Hungary
| |
Collapse
|
31
|
Gomaa EZ. Enhancement of γ-Amminobutyric Acid Production by Co-Culturing of Two Lactobacilli Strains. ACTA ACUST UNITED AC 2015. [DOI: 10.3923/ajbkr.2015.108.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Xiang Y, Liu H, Yan T, Zhuang Z, Jin D, Peng Y. Functional electrical stimulation-facilitated proliferation and regeneration of neural precursor cells in the brains of rats with cerebral infarction. Neural Regen Res 2014; 9:243-51. [PMID: 25206808 PMCID: PMC4146152 DOI: 10.4103/1673-5374.128215] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2013] [Indexed: 11/05/2022] Open
Abstract
Previous studies have shown that proliferation of endogenous neural precursor cells cannot alone compensate for the damage to neurons and axons. From the perspective of neural plasticity, we observed the effects of functional electrical stimulation treatment on endogenous neural precursor cell proliferation and expression of basic fibroblast growth factor and epidermal growth factor in the rat brain on the infarct side. Functional electrical stimulation was performed in rat models of acute middle cerebral artery occlusion. Simultaneously, we set up a placebo stimulation group and a sham-operated group. Immunohistochemical staining showed that, at 7 and 14 days, compared with the placebo group, the numbers of nestin (a neural precursor cell marker)-positive cells in the subgranular zone and subventricular zone were increased in the functional electrical stimulation treatment group. Western blot assays and reverse-transcription PCR showed that total protein levels and gene expression of epidermal growth factor and basic fibroblast growth factor were also upregulated on the infarct side. Prehensile traction test results showed that, at 14 days, prehension function of rats in the functional electrical stimulation group was significantly better than in the placebo group. These results suggest that functional electrical stimulation can promote endogenous neural precursor cell proliferation in the brains of acute cerebral infarction rats, enhance expression of basic fibroblast growth factor and epidermal growth factor, and improve the motor function of rats.
Collapse
Affiliation(s)
- Yun Xiang
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China ; Department of Rehabilitation Medicine, Shenzhen Sixth People's Hospital, Shenzhen, Guangdong Province, China
| | - Huihua Liu
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tiebin Yan
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhiqiang Zhuang
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Dongmei Jin
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan Peng
- Department of Rehabilitation Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong Province, China
| |
Collapse
|
33
|
Nava-Mesa MO, Jiménez-Díaz L, Yajeya J, Navarro-Lopez JD. GABAergic neurotransmission and new strategies of neuromodulation to compensate synaptic dysfunction in early stages of Alzheimer's disease. Front Cell Neurosci 2014; 8:167. [PMID: 24987334 PMCID: PMC4070063 DOI: 10.3389/fncel.2014.00167] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/02/2014] [Indexed: 01/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, brain atrophy due to neuronal and synapse loss, and formation of two pathological lesions: extracellular amyloid plaques, composed largely of amyloid-beta peptide (Aβ), and neurofibrillary tangles formed by intracellular aggregates of hyperphosphorylated tau protein. Lesions mainly accumulate in brain regions that modulate cognitive functions such as the hippocampus, septum or amygdala. These brain structures have dense reciprocal glutamatergic, cholinergic, and GABAergic connections and their relationships directly affect learning and memory processes, so they have been proposed as highly susceptible regions to suffer damage by Aβ during AD course. Last findings support the emerging concept that soluble Aβ peptides, inducing an initial stage of synaptic dysfunction which probably starts 20–30 years before the clinical onset of AD, can perturb the excitatory–inhibitory balance of neural circuitries. In turn, neurotransmission imbalance will result in altered network activity that might be responsible of cognitive deficits in AD. Therefore, Aβ interactions on neurotransmission systems in memory-related brain regions such as amygdaloid complex, medial septum or hippocampus are critical in cognitive functions and appear as a pivotal target for drug design to improve learning and dysfunctions that manifest with age. Since treatments based on glutamatergic and cholinergic pharmacology in AD have shown limited success, therapies combining modulators of different neurotransmission systems including recent findings regarding the GABAergic system, emerge as a more useful tool for the treatment, and overall prevention, of this dementia. In this review, focused on inhibitory systems, we will analyze pharmacological strategies to compensate neurotransmission imbalance that might be considered as potential therapeutic interventions in AD.
Collapse
Affiliation(s)
| | - Lydia Jiménez-Díaz
- Neurophysiology and Behavior Lab, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha Ciudad Real, Spain
| | - Javier Yajeya
- Department of Physiology and Pharmacology, University of Salamanca Salamanca, Spain
| | - Juan D Navarro-Lopez
- Neurophysiology and Behavior Lab, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha Ciudad Real, Spain
| |
Collapse
|
34
|
Arano T, Fujisaki S, Ikemoto MJ. Identification of tomoregulin-1 as a novel addicsin-associated factor. Neurochem Int 2014; 71:22-35. [DOI: 10.1016/j.neuint.2014.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 11/28/2022]
|
35
|
Rogers SK, Shapiro LA, Tobin RP, Tow B, Zuzek A, Mukherjee S, Newell-Rogers MK. Levetiracetam Differentially Alters CD95 Expression of Neuronal Cells and the Mitochondrial Membrane Potential of Immune and Neuronal Cells in vitro. Front Neurol 2014; 5:17. [PMID: 24600432 PMCID: PMC3927234 DOI: 10.3389/fneur.2014.00017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/29/2014] [Indexed: 01/09/2023] Open
Abstract
Epilepsy is a neurological seizure disorder that affects over 100 million people worldwide. Levetiracetam, either alone, as monotherapy, or as adjunctive treatment, is widely used to control certain types of seizures. Despite its increasing popularity as a relatively safe and effective anti-convulsive treatment option, its mechanism(s) of action are poorly understood. Studies have suggested neuronal, glial, and immune mechanisms of action. Understanding the precise mechanisms of action of levetiracetam would be extremely beneficial in helping to understand the processes involved in seizure generation and epilepsy. Moreover, a full understanding of these mechanisms would help to create more efficacious treatments while minimizing side-effects. The current study examined the effects of levetiracetam on the mitochondrial membrane potential of neuronal and non-neuronal cells, in vitro, in order to determine if levetiracetam influences metabolic processes in these cell types. In addition, this study sought to address possible immune-mediated mechanisms by determining if levetiracetam alters the expression of immune receptor–ligand pairs. The results show that levetiracetam induces expression of CD95 and CD178 on NGF-treated C17.2 neuronal cells. The results also show that levetiracetam increases mitochondrial membrane potential on C17.2 neuronal cells in the presence of nerve growth factor. In contrast, levetiracetam decreases the mitochondrial membrane potential of splenocytes and this effect was dependent on intact invariant chain, thus implicating immune cell interactions. These results suggest that both neuronal and non-neuronal anti-epileptic activities of levetiracetam involve control over energy metabolism, more specifically, mΔΨ. Future studies are needed to further investigate this potential mechanism of action.
Collapse
Affiliation(s)
| | - Lee A Shapiro
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA ; Central Texas Veterans Health Care System , Temple, TX , USA ; Scott and White Hospital , Temple, TX , USA
| | - Richard P Tobin
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA
| | - Benjamin Tow
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA
| | - Aleksej Zuzek
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA
| | - Sanjib Mukherjee
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA ; Central Texas Veterans Health Care System , Temple, TX , USA ; Scott and White Hospital , Temple, TX , USA
| | - M Karen Newell-Rogers
- Department of Surgery, Texas A&M University Health Science Center , Temple, TX , USA ; Scott and White Hospital , Temple, TX , USA
| |
Collapse
|
36
|
Griesmaier E, Stock K, Medek K, Stanika RI, Obermair GJ, Posod A, Wegleiter K, Urbanek M, Kiechl-Kohlendorfer U. Levetiracetam increases neonatal hypoxic-ischemic brain injury under normothermic, but not hypothermic conditions. Brain Res 2014; 1556:10-8. [PMID: 24530252 DOI: 10.1016/j.brainres.2014.01.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 01/20/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) resulting from perinatal asphyxia often leads to severe neurologic impairment or even death. There is a need to advance therapy for infants with HIE, for example to combine hypothermia with pharmacological treatment strategies. Levetiracetam (LEV) is approved for clinical administration to infants older than 4 weeks of age and is also used off-label in neonates. Furthermore, LEV was shown to be neuroprotective in adult animal models of brain injury. AIM OF THE STUDY The aim of this study was to evaluate the neuroprotective potential of LEV in vitro using primary hippocampal neurons, and in vivo using an established model of neonatal hypoxic-ischemic brain injury. RESULTS LEV treatment per se did not induce neurotoxicity in the developing rodent brain. Following oxygen glucose deprivation, we observed some, although not a significant, increase in cell death after LEV treatment. In vivo, LEV was administered under normothermic and hypothermic conditions following hypoxic-ischemic brain damage. LEV administration significantly increased brain injury under normothermic conditions. Compared to the normothermia-treated group, in the hypothermia group LEV administration did not increase hypoxic-ischemic brain injury. DISCUSSION This study demonstrates that LEV treatment increases neonatal hypoxic-ischemic brain injury. Administration of LEV in the acute phase of the injury might interfere with the balanced activation and inactivation of excitatory and inhibitory receptors in the developing brain. The neurotoxic effect of LEV in the injured newborn brain might further suggest an agonistic effect of LEV on the GABAergic system. Hypothermia treatment attenuates glutamate release following hypoxic-ischemic brain injury and might therefore limit the potentially deleterious effects of LEV. As a consequence, our findings do not necessarily rule out a potentially beneficial effect, but argue for cautious use of LEV in newborn infants with pre-existing brain injury.
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Pediatrics II, Innsbruck Medical University, Austria.
| | - Katharina Stock
- Department of Pediatrics II, Innsbruck Medical University, Austria
| | - Katharina Medek
- Department of Pediatrics II, Innsbruck Medical University, Austria; Division of Experimental Dermatology and Eb House, Paracelsus Medical University, Salzburg, Austria
| | - Ruslan I Stanika
- Department of Physiology and Medical Physics, Innsbruck Medical University, Austria
| | - Gerald J Obermair
- Department of Physiology and Medical Physics, Innsbruck Medical University, Austria
| | - Anna Posod
- Department of Pediatrics II, Innsbruck Medical University, Austria
| | - Karina Wegleiter
- Department of Pediatrics II, Innsbruck Medical University, Austria
| | - Martina Urbanek
- Department of Pediatrics II, Innsbruck Medical University, Austria
| | | |
Collapse
|
37
|
Al-Shorbagy MY, El Sayeh BM, Abdallah DM. Additional antiepileptic mechanisms of levetiracetam in lithium-pilocarpine treated rats. PLoS One 2013; 8:e76735. [PMID: 24098559 PMCID: PMC3789684 DOI: 10.1371/journal.pone.0076735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 08/28/2013] [Indexed: 01/22/2023] Open
Abstract
Several studies have addressed the antiepileptic mechanisms of levetiracetam (LEV); however, its effect on catecholamines and the inflammatory mediators that play a role in epilepsy remain elusive. In the current work, lithium (Li) pretreated animals were administered LEV (500 mg/kg i.p) 30 min before the induction of convulsions by pilocarpine (PIL). Li-PIL-induced seizures were accompanied by increased levels of hippocampal prostaglandin (PG) E2, myeloperoxidase (MPO), tumor necrosis factor-α, and interleukin-10. Moreover, it markedly elevated hippocampal lipid peroxides and nitric oxide levels, while it inhibited the glutathione content. Li-PIL also reduced hippocampal noradrenaline, as well as dopamine contents. Pretreatment with LEV protected against Li-PIL-induced seizures, where it suppressed the severity and delayed the onset of seizures in Li-PIL treated rats. Moreover, LEV reduced PGE2 and MPO, yet it did not affect the level of both cytokines in the hippocampus. LEV also normalized hippocampal noradrenaline, dopamine, glutathione, lipid peroxides, and nitric oxide contents. In conclusion, alongside its antioxidant property, LEV anticonvulsive effect involves catecholamines restoration, as well as inhibition of PGE2, MPO, and nitric oxide.
Collapse
Affiliation(s)
- Muhammad Y. Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Bahia M. El Sayeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalaal M. Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
38
|
Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, Smith SB, Ganapathy V, Maher P. The cystine/glutamate antiporter system x(c)(-) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal 2013; 18:522-55. [PMID: 22667998 PMCID: PMC3545354 DOI: 10.1089/ars.2011.4391] [Citation(s) in RCA: 682] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The antiporter system x(c)(-) imports the amino acid cystine, the oxidized form of cysteine, into cells with a 1:1 counter-transport of glutamate. It is composed of a light chain, xCT, and a heavy chain, 4F2 heavy chain (4F2hc), and, thus, belongs to the family of heterodimeric amino acid transporters. Cysteine is the rate-limiting substrate for the important antioxidant glutathione (GSH) and, along with cystine, it also forms a key redox couple on its own. Glutamate is a major neurotransmitter in the central nervous system (CNS). By phylogenetic analysis, we show that system x(c)(-) is a rather evolutionarily new amino acid transport system. In addition, we summarize the current knowledge regarding the molecular mechanisms that regulate system x(c)(-), including the transcriptional regulation of the xCT light chain, posttranscriptional mechanisms, and pharmacological inhibitors of system x(c)(-). Moreover, the roles of system x(c)(-) in regulating GSH levels, the redox state of the extracellular cystine/cysteine redox couple, and extracellular glutamate levels are discussed. In vitro, glutamate-mediated system x(c)(-) inhibition leads to neuronal cell death, a paradigm called oxidative glutamate toxicity, which has successfully been used to identify neuroprotective compounds. In vivo, xCT has a rather restricted expression pattern with the highest levels in the CNS and parts of the immune system. System x(c)(-) is also present in the eye. Moreover, an elevated expression of xCT has been reported in cancer. We highlight the diverse roles of system x(c)(-) in the regulation of the immune response, in various aspects of cancer and in the eye and the CNS.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sanchez PE, Zhu L, Verret L, Vossel KA, Orr AG, Cirrito JR, Devidze N, Ho K, Yu GQ, Palop JJ, Mucke L. Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer's disease model. Proc Natl Acad Sci U S A 2012; 109:E2895-903. [PMID: 22869752 PMCID: PMC3479491 DOI: 10.1073/pnas.1121081109] [Citation(s) in RCA: 472] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In light of the rising prevalence of Alzheimer's disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-β peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-β-induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.
Collapse
Affiliation(s)
- Pascal E. Sanchez
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - Lei Zhu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - Laure Verret
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - Keith A. Vossel
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - Anna G. Orr
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - John R. Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Nino Devidze
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Jorge J. Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology, University of California, San Francisco, CA 94158; and
| |
Collapse
|
40
|
Fukuyama K, Tanahashi S, Nakagawa M, Yamamura S, Motomura E, Shiroyama T, Tanii H, Okada M. Levetiracetam inhibits neurotransmitter release associated with CICR. Neurosci Lett 2012; 518:69-74. [DOI: 10.1016/j.neulet.2012.03.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 11/27/2022]
|
41
|
Héja L, Nyitrai G, Kékesi O, Dobolyi A, Szabó P, Fiáth R, Ulbert I, Pál-Szenthe B, Palkovits M, Kardos J. Astrocytes convert network excitation to tonic inhibition of neurons. BMC Biol 2012; 10:26. [PMID: 22420899 PMCID: PMC3342137 DOI: 10.1186/1741-7007-10-26] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/15/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Glutamate and γ-aminobutyric acid (GABA) transporters play important roles in balancing excitatory and inhibitory signals in the brain. Increasing evidence suggest that they may act concertedly to regulate extracellular levels of the neurotransmitters. RESULTS Here we present evidence that glutamate uptake-induced release of GABA from astrocytes has a direct impact on the excitability of pyramidal neurons in the hippocampus. We demonstrate that GABA, synthesized from the polyamine putrescine, is released from astrocytes by the reverse action of glial GABA transporter (GAT) subtypes GAT-2 or GAT-3. GABA release can be prevented by blocking glutamate uptake with the non-transportable inhibitor DHK, confirming that it is the glutamate transporter activity that triggers the reversal of GABA transporters, conceivably by elevating the intracellular Na+ concentration in astrocytes. The released GABA significantly contributes to the tonic inhibition of neurons in a network activity-dependent manner. Blockade of the Glu/GABA exchange mechanism increases the duration of seizure-like events in the low-[Mg2+] in vitro model of epilepsy. Under in vivo conditions the increased GABA release modulates the power of gamma range oscillation in the CA1 region, suggesting that the Glu/GABA exchange mechanism is also functioning in the intact hippocampus under physiological conditions. CONCLUSIONS The results suggest the existence of a novel molecular mechanism by which astrocytes transform glutamatergic excitation into GABAergic inhibition providing an adjustable, in situ negative feedback on the excitability of neurons.
Collapse
Affiliation(s)
- László Héja
- Department of Functional Pharmacology, Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Pusztaszeri 59-67, 1025 Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hanaya R, Kiura Y, Serikawa T, Kurisu K, Arita K, Sasa M. Modulation of abnormal synaptic transmission in hippocampal CA3 neurons of spontaneously epileptic rats (SERs) by levetiracetam. Brain Res Bull 2011; 86:334-9. [PMID: 21968023 DOI: 10.1016/j.brainresbull.2011.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 09/17/2011] [Accepted: 09/19/2011] [Indexed: 11/18/2022]
Abstract
Levetiracetam (LEV) inhibits partial refractory epilepsy in human, and both convulsive and absence-like seizures in the spontaneously epileptic rat (SER). Two-thirds of hippocampal CA3 neurons in SER show a long-lasting depolarization shift, with accompanying repetitive firing upon mossy fiber stimulation. This abnormal excitability is probably attributable to abnormalities in the L-type Ca(2+) channels. We performed electrophysiological studies to elucidate the mechanism underlying the antiepileptic effects of LEV via intracellular recording from the hippocampal CA3 neurons in slice preparations of SER and non-epileptic Wistar rats. LEV (100 μM) inhibited the depolarization shift with repetitive firing by mossy fiber stimulation (MFS), without affecting the first spike in SER CA3 neurons. At a higher dose (1mM), LEV suppressed the first spike in all SER neurons (including the CA3 neurons which showed only a single action potential by MFS), while the single action potential of Wistar rat CA3 neurons remained unaffected. SER CA3 neurons with MFS-induced abnormal firing exhibited a higher number of repetitive spikes when a depolarization pulse was applied in the SER CA3 neurons. LEV (100 μM, 1mM) reduced the repetitive firing induced by a depolarization pulse applied without affecting Ca(2+) spike in SER neurons. LEV is known not to bind glutamate and gamma-aminobutyric acid (GABA) receptors. These findings suggest that the therapeutic concentration of LEV inhibits abnormal firing of the CA3 neurons by modulating abnormal synaptic transmission and abnormal Na(+) channels in SER.
Collapse
Affiliation(s)
- Ryosuke Hanaya
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Ueda Y, Kitamoto A, Willmore LJ, Kojima T. Hippocampal gene network analysis in an experimental model of posttraumatic epilepsy. Neurochem Res 2010; 36:1323-8. [PMID: 21191651 DOI: 10.1007/s11064-010-0386-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2010] [Indexed: 11/25/2022]
Abstract
In the present study, we performed comprehensive gene expression and gene network analyses using a DNA microarray to elucidate the molecular events responsible for the pathology of posttraumatic epilepsy at the partial seizure stage. We used an experimental posttraumatic epilepsy model of amygdalar focal FeCl(3)-injected rats and compared gene expression profiles in the hippocampus at the partial seizure stage (less than stage 3 on Racine's convulsion scale) and that of sham-operated animals. At the partial seizure stage, upregulation of phospholipase A2 (PLA2) and lipid metabolism were observed, which have been reported to be caused by brain injury and seizures in previous studies. Furthermore, significant upregulation of genes related to inflammation and the immune system was observed. These molecular changes in PLA2 and lipid metabolism may be related to seizure propagation.
Collapse
Affiliation(s)
- Yuto Ueda
- Section of Psychiatry, Department of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | | | | | | |
Collapse
|
44
|
Micov A, Tomić M, Popović B, Stepanović-Petrović R. The antihyperalgesic effect of levetiracetam in an inflammatory model of pain in rats: mechanism of action. Br J Pharmacol 2010; 161:384-92. [PMID: 20735422 PMCID: PMC2989589 DOI: 10.1111/j.1476-5381.2010.00877.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 04/09/2010] [Accepted: 04/12/2010] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Levetiracetam, a novel antiepileptic drug, has recently been shown to have antinociceptive effects in various animal models of pain. The purpose of this study was to investigate the antihyperalgesic effect of levetiracetam and its mechanism of action, by examining the involvement of GABAergic, opioidergic, 5-hydroxytryptaminergic (5-HTergic) and adrenergic systems in its effect, in a rat model of inflammatory pain. EXPERIMENTAL APPROACH Rats were intraplantarly injected with the pro-inflammatory compound carrageenan. A paw pressure test was used to determine: (i) the effect of levetiracetam on carrageenan-induced hyperalgesia; and (ii) the effects of bicuculline (selective GABA(A) receptor antagonist), naloxone (non-selective opioid receptor antagonist), methysergide (non-selective 5-HT receptor antagonist) and yohimbine (selective alpha(2)-adrenoceptor antagonist) on the antihyperalgesic action of levetiracetam. RESULTS Levetiracetam (10-200 mg.kg(-1); p.o.) significantly reduced, in a dose-dependent manner, the inflammatory hyperalgesia induced by carrageenan. The antihyperalgesic effect of levetiracetam was significantly decreased after administration of bicuculline (0.5-2 mg.kg(-1); i.p.), naloxone (1-3 mg.kg(-1); i.p.), methysergide (0.25-1 mg.kg(-1); i.p.) and yohimbine (1-3 mg.kg(-1); i.p.). CONCLUSIONS AND IMPLICATIONS These results show that levetiracetam produced antihyperalgesia which is at least in part mediated by GABA(A), opioid, 5-HT and alpha(2)-adrenergic receptors, in an inflammatory model of pain. The efficacy of levetiracetam in this animal model of inflammatory pain suggests that it could be a potentially important agent for treating inflammatory pain conditions in humans.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Carrageenan
- Disease Models, Animal
- Hyperalgesia/drug therapy
- Hyperalgesia/immunology
- Hyperalgesia/metabolism
- Levetiracetam
- Male
- Narcotic Antagonists
- Pain/drug therapy
- Pain/immunology
- Pain/metabolism
- Piracetam/administration & dosage
- Piracetam/analogs & derivatives
- Piracetam/pharmacology
- Piracetam/therapeutic use
- Rats
- Rats, Wistar
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, GABA-A/metabolism
- Receptors, Opioid/metabolism
- Receptors, Serotonin/metabolism
Collapse
Affiliation(s)
- A Micov
- Department of Pharmacology, Faculty of Pharmacy, University of BelgradeBelgrade, Serbia
| | - M Tomić
- Department of Pharmacology, Faculty of Pharmacy, University of BelgradeBelgrade, Serbia
| | - B Popović
- Statistical Office of the Republic of SerbiaBelgrade, Serbia
| | - R Stepanović-Petrović
- Department of Pharmacology, Faculty of Pharmacy, University of BelgradeBelgrade, Serbia
| |
Collapse
|
45
|
Brown ES. Effects of glucocorticoids on mood, memory, and the hippocampus. Treatment and preventive therapy. Ann N Y Acad Sci 2009; 1179:41-55. [PMID: 19906231 DOI: 10.1111/j.1749-6632.2009.04981.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Corticosteroids, such as prednisone and dexamethasone, are commonly prescribed medications that suppress the immune system and decrease inflammation. Common side effects of long-term treatment with corticosteroids include weight gain, osteoporosis, and diabetes mellitus. This paper reviews the literature on psychiatric and cognitive changes during corticosteroid therapy and potential treatment options. Hypomania and mania are the most common mood changes during acute corticosteroid therapy, although depression has also been reported. However, depression is reported to be more common than mania during long-term treatment with corticosteroids. A decline in declarative and working memory is also reported during corticosteroid therapy. Corticosteroids are associated with changes in the temporal lobe, detected by structural, functional, and spectroscopic imaging. The mood and cognitive symptoms are dose dependent and frequently occur during the first few weeks of therapy. Other risk factors are not well characterized. Controlled trials suggest that lithium and phenytoin can prevent mood symptoms associated with corticosteroids. Lamotrigine and memantine also have been shown to reverse, at least partially, the declarative memory effects of corticosteroids. Uncontrolled trials suggest that antipsychotics, anti-seizure medications, and perhaps some antidepressants can also be useful for normalizing mood changes associated with corticosteroids. Thus, both the symptoms and treatment response are similar to those of bipolar disorder. Moreover, corticosteroid-induced mood and cognitive alterations have been shown to be reversible with dose reduction or discontinuation of treatment.
Collapse
Affiliation(s)
- E Sherwood Brown
- Psychoneuroendocrine Research Program, Department of Psychiatry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
| |
Collapse
|
46
|
Héja L, Barabás P, Nyitrai G, Kékesi KA, Lasztóczi B, Toke O, Tárkányi G, Madsen K, Schousboe A, Dobolyi A, Palkovits M, Kardos J. Glutamate uptake triggers transporter-mediated GABA release from astrocytes. PLoS One 2009; 4:e7153. [PMID: 19777062 PMCID: PMC2744931 DOI: 10.1371/journal.pone.0007153] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 09/02/2009] [Indexed: 11/21/2022] Open
Abstract
Background Glutamate (Glu) and γ-aminobutyric acid (GABA) transporters play important roles in regulating neuronal activity. Glu is removed from the extracellular space dominantly by glial transporters. In contrast, GABA is mainly taken up by neurons. However, the glial GABA transporter subtypes share their localization with the Glu transporters and their expression is confined to the same subpopulation of astrocytes, raising the possibility of cooperation between Glu and GABA transport processes. Methodology/Principal Findings Here we used diverse biological models both in vitro and in vivo to explore the interplay between these processes. We found that removal of Glu by astrocytic transporters triggers an elevation in the extracellular level of GABA. This coupling between excitatory and inhibitory signaling was found to be independent of Glu receptor-mediated depolarization, external presence of Ca2+ and glutamate decarboxylase activity. It was abolished in the presence of non-transportable blockers of glial Glu or GABA transporters, suggesting that the concerted action of these transporters underlies the process. Conclusions/Significance Our results suggest that activation of Glu transporters results in GABA release through reversal of glial GABA transporters. This transporter-mediated interplay represents a direct link between inhibitory and excitatory neurotransmission and may function as a negative feedback combating intense excitation in pathological conditions such as epilepsy or ischemia.
Collapse
Affiliation(s)
- László Héja
- Department of Neurochemistry, Institute of Biomolecular Chemistry, Chemical Research Center, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ueda Y, Doi T, Takaki M, Nagatomo K, Nakajima A, Willmore LJ. Levetiracetam enhances endogenous antioxidant in the hippocampus of rats: In vivo evaluation by brain microdialysis combined with ESR spectroscopy. Brain Res 2009; 1266:1-7. [DOI: 10.1016/j.brainres.2009.02.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 01/05/2009] [Accepted: 02/21/2009] [Indexed: 10/21/2022]
|
48
|
Trinka E, Dobesberger J. New treatment options in status epilepticus: a critical review on intravenous levetiracetam. Ther Adv Neurol Disord 2009; 2:79-91. [PMID: 21180643 PMCID: PMC3002622 DOI: 10.1177/1756285608100460] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The effectiveness of Levetiracetam (LEV) in the treatment of focal and generalised epilepsies is well established. LEV has a wide spectrum of action, good tolerability and a favourable pharmacokinetic profile. An injectable formulation has been released as an intravenous (IV) infusion in 2006 for patients with epilepsy when oral administration is temporarily not feasible. Bioequivalence to the oral preparation has been demonstrated with good tolerability and safety enabling a smooth transition from oral to parenteral formulation and vice versa. Although IV LEV is not licensed for treatment of status epilepticus (SE), open-label experience in retrospective case series is accumulating. Until now (August 2008) 156 patients who were treated with IV LEV for various forms of SE have been reported with an overall success rate of 65.4%. The most often used initial dose was 2000-3000 mg over 15 minutes. Adverse events were reported in 7.1%, and were mild and transient. Although IV LEV is an interesting alternative for the treatment of SE due to the lack of centrally depressive effects and low potential of drug interactions, one has to be aware of the nonrandomised retrospective study design, the heterogenous patient population and treatment protocols, and the publication bias inherent in these type of studies. Only a large randomised controlled trial with an adequate comparator will reveal the efficacy and effectiveness of this promising new IV formulation.
Collapse
Affiliation(s)
- Eugen Trinka
- Medical University Innsbruck, Department of Neurology, Innsbruck, Austria
| | | |
Collapse
|
49
|
Perdan K, Lipnik‐Štangelj M, Kržan M. Chapter 8 The Impact of Astrocytes in the Clearance of Neurotransmitters by Uptake and Inactivation. ADVANCES IN PLANAR LIPID BILAYERS AND LIPOSOMES 2009. [DOI: 10.1016/s1554-4516(09)09008-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
50
|
Levetiracetam in stiff-person syndrome. J Neurol 2008; 255:1721-5. [DOI: 10.1007/s00415-008-0007-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 04/23/2008] [Accepted: 05/20/2008] [Indexed: 11/25/2022]
|