1
|
Mohammed HS, Ahmed DH, Khadrawy YA, Madian NG. Neuroprotection in pentylenetetrazol kindling rat model: A synergistic approach with eugenol and photobiomodulation. Brain Res 2025; 1858:149645. [PMID: 40228569 DOI: 10.1016/j.brainres.2025.149645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/21/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures, significantly impacting patient health and quality of life. This study explores the neuroprotective effects of combining Eugenol (EUG), a natural bioactive compound administered at 100 mg/kg, with photobiomodulation (PBM), a non-invasive low-level laser therapy at 830 nm wavelength and 100 mW power, in a pentylenetetrazole (PTZ) kindling rat model of epilepsy. Fifty-nine adult male Wistar rats were assigned to five experimental groups: Control, PTZ (epilepsy model), PBM, EUG, and EUG + PBM. Seizure severity was assessed using a modified Racine scale following each PTZ injection. The study also evaluated cortical and hippocampal levels of brain-derived neurotrophic factor (BDNF), oxidative stress markers (MDA, NO, and GSH), activities of acetylcholinesterase (AChE) and Na + K + -ATPase, and monoamine neurotransmitters (DA, 5-HT, and NE). The results demonstrated that EUG and PBM, both individually and combined, significantly reduced seizure severity, mitigated oxidative stress, restored enzyme activities, and elevated BDNF levels. The combined treatment yielded superior neuroprotective effects compared to individual interventions, emphasizing its potential as a promising therapeutic strategy for epilepsy management.
Collapse
Affiliation(s)
- Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Dalia H Ahmed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, National Research Center, Giza, Egypt
| | - Noha G Madian
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Cheng CJ, Wang LC, Chu LJ, Chen KY, Huang CY, Lan KL, Huang KY. Extracellular vesicles from fifth-stage larval Angiostrongylus cantonensis upregulate cholesterol biosynthesis and suppress NLRP2-associated inflammatory responses in mouse astrocytes. mSystems 2025; 10:e0101424. [PMID: 39636121 PMCID: PMC11748502 DOI: 10.1128/msystems.01014-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Angiostrongylus cantonensis is a zoonotic parasite that causes severe symptoms in humans, including eosinophilic meningitis and eosinophilic meningoencephalitis. Extracellular vesicles (EVs) derived from helminthes have been implicated in regulating host survival and immune response. However, the roles of A. cantonensis EVs in modulating parasite pathogenesis and host immune response remain poorly understood. Herein, we characterized EVs derived from A. cantonensis fifth-stage larvae (L5) and adult worms. Ultrastructural features showed that EVs from adult worms are smaller in size compared with those from L5. Proteomic analysis identified stage-specific proteins packaged in L5 and adult worm EVs. To investigate the crosstalk between L5 EVs and host cells, RNA sequencing analysis was conducted to identify the differentially expressed genes (DEGs) and enriched biological pathways in mouse astrocytes treated with L5 EVs. GO and KEGG enrichment analysis demonstrated that the pathways related to "cholesterol biosynthesis" are significantly upregulated in L5 EV-treated astrocytes. Based on the transcriptomic data, we observed a downregulated trend of NOD-like receptors (NLRs) protein 2 (NLRP2), a key regulator of brain inflammation, in mouse astrocytes treated with L5 EVs. To validate this result, we utilized ATP to induce the expression of NLRP2 inflammasome-related genes and proteins, as well as the secretion of downstream cytokines. Notably, ATP-induced overexpression of NLRP2 inflammasome-related molecules was significantly reduced in mouse astrocytes upon L5 EV treatment. Collectively, our data suggest that A. cantonensis L5 EVs enhance cholesterol synthesis and potentially modulate immune response by reducing NLRP2 inflammasome-related signaling in non-permissive host cells.IMPORTANCEAngiostrongylus cantonensis is a significant causative agent of eosinophilic meningitis and eosinophilic meningoencephalitis in humans. Helminth-derived extracellular vesicles (EVs) are known to play a crucial role in parasite pathogenesis and host immunomodulation. However, the protein compositions of A. cantonensis EVs and their roles in parasite pathogenesis and host immune response remain unclear. Our results demonstrate for the first time the distinct protein compositions in A. cantonensis L5 and adult worm EVs. The highly abundant proteins in L5 EVs that have immunomodulatory or pathogenic potential in the host deserve further investigation. Additionally, the uptake of L5 EVs by mouse astrocytes significantly upregulates cholesterol synthesis and suppresses ATP-induced NLRP2 inflammasome-related signaling. This study highlights the immunomodulatory roles of L5 EVs in non-permissive hosts, suggesting their potential as therapeutic targets and vaccine candidates against A. cantonensis.
Collapse
Affiliation(s)
- Chien-Ju Cheng
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Lichieh Julie Chu
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
- Department of Otolaryngology—Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Kuang-Yao Chen
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| | - Ching-Yun Huang
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Lun Lan
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, Taiwan
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
3
|
Nazmy MM, Noor NA, Mohammed FF, Khadrawy YA, Radwan NM. Taurine protection attenuates bisphenol-A-induced behavioral, neurochemical, and histopathological alterations in male rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03767-4. [PMID: 39820545 DOI: 10.1007/s00210-024-03767-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/25/2024] [Indexed: 01/19/2025]
Abstract
Due to the continuous exposure to bisphenol-A (BPA), the current study was conducted to evaluate taurine's neuroprotective action against BPA's adverse effect on the brain. Rats were grouped into control, BPA-treated rats, and taurine + BPA-treated rats. At the end of the 35-day treatment period, the memory of the rats was evaluated using the novel object test and the Y-maze test. An open-field test was used to measure motor activity. The changes in monoamines, monoamine oxidase (MAO), acetylcholinesterase (AChE), Na+,K+,ATPase, oxidative stress, caspase-3, and histopathology were evaluated in the cortical and hippocampal tissues of all groups. Data analysis by ANOVA revealed that BPA treatment induced motor hyperactivity and short- and long-term memory impairment. In the cortex, BPA decreased serotonin (5-HT), norepinephrine (NE), MAO, Na+,K+,ATPase, and nitric oxide (NO) and increased dopamine (DA), AChE, lipid peroxidation (MDA), glutathione (GSH), and caspase-3. In the hippocampus, BPA increased 5-HT, DA, NE, MAO, AChE, MDA, NO, GSH, and caspase-3 and decreased Na+,K+,ATPase. These neurochemical changes were accompanied by significant histopathological alterations. Taurine treatment prevented memory impairment and motor hyperactivity induced by BPA. Taurine attenuated the neurochemical changes, oxidative stress, and caspase-3 level. Taurine improved the histopathological change induced by BPA. In conclusion, taurine significantly prevented BPA-induced cognitive deficits, motor coordination impairments, neurotransmitter imbalances, histopathological alterations, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Mohamed M Nazmy
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University, Badr City, Cairo, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Neveen A Noor
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Faten F Mohammed
- Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Department of Pathology, College of Veterinary Medicine, King Faisal University, 31982, Al-Ahsa, Saudi Arabia
| | - Yasser A Khadrawy
- Medical Physiology Department, Clinical Studies and Medical Research Institute, National Research Centre, Giza, Egypt.
| | - Nasr M Radwan
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
4
|
Recart VM, Spohr L, de Aguiar MSS, de Souza AA, Goularte KCM, Bona NP, Pedra NS, Teixeira FC, Stefanello FM, Spanevello RM. Gallic acid attenuates lipopolysaccharide - induced memory deficits, neurochemical changes, and peripheral alterations in purinergic signaling. Metab Brain Dis 2024; 40:43. [PMID: 39601942 DOI: 10.1007/s11011-024-01424-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/14/2024] [Indexed: 11/29/2024]
Abstract
Neuroinflammation is associated with many neurological disorders. Gallic acid (GA) has attracted significant attention due to its biological properties, such as neuroprotective, anti-inflammatory, and antioxidant effects. In this study, we evaluated the effects of GA in memory, TNF-α levels, oxidative stress, and activities of acetylcholinesterase (AChE), Na+, K+-ATPase and Ca2+-ATPase in the brain of mice exposed to lipopolysaccharide (LPS). Additionally, we evaluated alterations in adenine nucleotides and nucleosides in the serum. Male mice were orally pretreated with vehicle or GA (50 or 100 mg/kg) for 14 days. Between days 8 and 14, the animals also received LPS injection (250 µg/kg) or saline. At the end of the experimental protocol, the animals were submitted to object recognition test, euthanized and cerebral cortex, hippocampus, striatum and blood were collected. LPS induced memory deficits, which were prevented by GA treatment. GA protected against LPS-induced oxidative damage in the cerebral cortex, hippocampus and striatum by reducing reactive oxygen species and nitrite levels, while increasing total thiol content and activities of antioxidant enzymes. GA also prevented LPS-induced alterations in AChE, Na+, K+-ATPase, and Ca2+-ATPase activities in brain structures. LPS elevated TNF-α levels in the hippocampus and cerebral cortex, which were attenuated by GA treatment. Furthermore, LPS caused a reduction in ADP and AMP hydrolysis and an increase in adenosine deamination in the serum, which were also prevented by GA. The effects of GA against neuroinflammation may be attributed to its potent antioxidant and anti-inflammatory properties, which modulate various pathways, including those involved in memory mechanisms.
Collapse
Affiliation(s)
- Vânia Machado Recart
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Anita Avila de Souza
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Kelen Cristiane Machado Goularte
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Fernanda Cardoso Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Capão do Leão, RS, Brazil.
| |
Collapse
|
5
|
Singh PK, Shekh-Ahmad T. Nrf2 as a potential target for the treatment of epilepsy. Neural Regen Res 2024; 19:1865-1866. [PMID: 38227501 DOI: 10.4103/1673-5374.390975] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/22/2023] [Indexed: 01/17/2024] Open
Affiliation(s)
- Prince Kumar Singh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
6
|
Sutha J, Gayathri M, Ramesh M. Chronic exposure to tris (2-chloroethyl) phosphate (TCEP) induces brain structural and functional changes in zebrafish (Danio rerio): A comparative study on the environmental and LC50 concentrations of TCEP. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:16770-16781. [PMID: 38321284 DOI: 10.1007/s11356-024-32154-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
Tris (2-chloroethyl) phosphate (TCEP) is a crucial organophosphorus flame retardant widely used in many industrial and commercial products. Available reports reported that TCEP could cause various toxicological effects on organisms, including humans. Unfortunately, toxicity data for TCEP (particularly on neurotoxicity) on aquatic organisms are lacking. In the present study, Danio rerio were exposed to different concentrations of TCEP for 42 days (chronic exposure), and oxidative stress, neurotoxicity, sodium, potassium-adenosine triphosphatase (Na+, K+-ATPase) activity, and histopathological changes were evaluated in the brain. The results showed that TCEP (100 and 1500 µg L-1) induced oxidative stress and significantly decreased the activities of antioxidant enzymes (SOD, CAT and GR) in the brain tissue of zebrafish. In contrast, the lipid peroxidation (LPO) level was increased compared to the control group. Exposure to TCEP inhibited the acetylcholinesterase (AChE) and Na+,K+-ATPase activities in the brain tissue. Brain histopathology after 42 days of exposure to TCEP showed cytoplasmic vacuolation, inflammatory cell infiltration, degenerated neurons, degenerated purkinje cells and binucleate. Furthermore, TCEP exposure leads to significant changes in dopamine and 5-HT levels in the brain of zebrafish. The data in the present study suggest that high concentrations of TCEP might affect the fish by altering oxidative balance and inducing marked pathological changes in the brain of zebrafish. These findings indicate that chronic exposure to TCEP may cause a neurotoxic effect in zebrafish.
Collapse
Affiliation(s)
- Jesudass Sutha
- Unit of Toxicology, Department of Zoology, School of Life Sciences, Bharathiar University, 641 046, Coimbatore, Tamil Nadu, India
| | - Murugesh Gayathri
- Unit of Toxicology, Department of Zoology, School of Life Sciences, Bharathiar University, 641 046, Coimbatore, Tamil Nadu, India
| | - Mathan Ramesh
- Unit of Toxicology, Department of Zoology, School of Life Sciences, Bharathiar University, 641 046, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
7
|
Crespo M, León-Navarro DA, Martín M. Maternal Caffeine Consumption during Gestation and Lactation Abolishes Cortical Oxidative Stress and Restores Na +/K +-ATPase Activity in Neonates Exposed to Hyperthermia-Induced Seizures. Biomedicines 2023; 11:3292. [PMID: 38137513 PMCID: PMC10741554 DOI: 10.3390/biomedicines11123292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Caffeine is a psychoactive substance that is widely consumed by individuals of various demographics, including pregnant women. It can readily cross the blood-brain and placental barriers, easily reaching the fetal brain. In addition, caffeine has also shown antioxidant properties, as its consumption reduces oxidative stress in various pathologies, including epilepsy. Febrile seizures (FS) are among the most common convulsive disorders in infants and young children. Here, we used an animal model of FS to learn whether maternal caffeine (1 g/L) intake consumption during gestation and lactation could exert beneficial effects on the rat cortex. Neonatal development was analyzed by measuring pinna opening, eye opening, righting reflex on the surface, and geotaxis reflex. Five and twenty days after HIS, the rats were euthanized, and plasma membranes and cytosolic fractions were isolated from their cortex brain. The enzymatic activities of glutathione reductase, glutathione S-transferase, Na+/K+-ATPase, and Mg2+-ATPase, as well as the levels of thiobarbituric acid reacting substances, were quantified. Results showed that maternal caffeine intake eliminates oxidative stress and normalizes Na+/K+-ATPase activity disrupted by HIS and also affects some parameters relating to the neurodevelopment of neonates. As FS in infants has been related to epilepsy in adults, the antioxidant properties of caffeine could prevent potential damage from hyperthermia.
Collapse
Affiliation(s)
- María Crespo
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - David Agustín León-Navarro
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Mairena Martín
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain;
| |
Collapse
|
8
|
Madireddy S, Madireddy S. Therapeutic Strategies to Ameliorate Neuronal Damage in Epilepsy by Regulating Oxidative Stress, Mitochondrial Dysfunction, and Neuroinflammation. Brain Sci 2023; 13:brainsci13050784. [PMID: 37239256 DOI: 10.3390/brainsci13050784] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Epilepsy is a central nervous system disorder involving spontaneous and recurring seizures that affects 50 million individuals globally. Because approximately one-third of patients with epilepsy do not respond to drug therapy, the development of new therapeutic strategies against epilepsy could be beneficial. Oxidative stress and mitochondrial dysfunction are frequently observed in epilepsy. Additionally, neuroinflammation is increasingly understood to contribute to the pathogenesis of epilepsy. Mitochondrial dysfunction is also recognized for its contributions to neuronal excitability and apoptosis, which can lead to neuronal loss in epilepsy. This review focuses on the roles of oxidative damage, mitochondrial dysfunction, NAPDH oxidase, the blood-brain barrier, excitotoxicity, and neuroinflammation in the development of epilepsy. We also review the therapies used to treat epilepsy and prevent seizures, including anti-seizure medications, anti-epileptic drugs, anti-inflammatory therapies, and antioxidant therapies. In addition, we review the use of neuromodulation and surgery in the treatment of epilepsy. Finally, we present the role of dietary and nutritional strategies in the management of epilepsy, including the ketogenic diet and the intake of vitamins, polyphenols, and flavonoids. By reviewing available interventions and research on the pathophysiology of epilepsy, this review points to areas of further development for therapies that can manage epilepsy.
Collapse
Affiliation(s)
- Sahithi Madireddy
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
9
|
Bilister Egilmez C, Azak Pazarlar B, Erdogan MA, Erbas O. N-acetyl cysteine: A new look at its effect on PTZ-induced convulsions. Epilepsy Res 2023; 193:107144. [PMID: 37116249 DOI: 10.1016/j.eplepsyres.2023.107144] [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: 10/25/2022] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
INTRODUCTION/AIM Epilepsy is widely investigated as a common neurological disease requiring pharmacologically effective agents. N-acetyl cysteine (NAC), has become a remarkable molecule with its role in both antioxidant and glutaminergic modulation. There are many points and processes waiting to be revealed regarding the role of NAC in epilepsy. MATERIALS AND METHODS Pentylenetetrazole (PTZ) was administered to induce seizures in a total number of 48 Sprague-Dawley rats. 35 mg/kg PTZ dose as a sub-convulsive dose was administered to 24 animals to monitor EEG changes, while 70 mg/kg PTZ dose which was a convulsive dose was administered to 24 animals to determine seizure-related behavioral changes with the Racine's scale. 30 min before the seizure-induced procedure, NAC was administered at doses of 300 and 600 mg/kg as pretreatment to investigate anti-seizure and anti-oxidative effects. The spike percentage, the stage of convulsion, and the onset time of the first myoclonic jerk were evaluated to determine the anti-seizure effect. Furthermore, its effect on oxidative stress was determined by measuring both malondialdehyde (MDA) level and superoxide dismutase (SOD) enzyme activity. RESULTS There was a dose-dependent reduction in the seizure stage and prolonged onset time of the first myoclonic jerk in rats with NAC pretreatment. EEG recordings resulted in a dose-dependent decrease in spike percentages. Moreover, the same dose-dependent changes were observed in oxidative stress biomarkers, both 300 mg/kg NAC and 600 mg/kg decreased MDA levels and ameliorated SOD activity. CONCLUSION We can report that 300 mg/kg and 600 mg/kg doses of NAC are promising with their reducing effect on convulsions and have a beneficial effect by preventing oxidative stress. In addition, NAC has been also determined that this effect is dose-dependent. Detailed and comparative studies are needed on the convulsion-reducing effect of NAC in epilepsy.
Collapse
Affiliation(s)
- Cansu Bilister Egilmez
- Faculty of Medicine, Department of Physiology, Izmir Katip Celebi University, Izmir, Turkey.
| | - Burcu Azak Pazarlar
- Faculty of Medicine, Department of Physiology, Izmir Katip Celebi University, Izmir, Turkey
| | - Mumin Alper Erdogan
- Faculty of Medicine, Department of Physiology, Izmir Katip Celebi University, Izmir, Turkey
| | - Oytun Erbas
- Faculty of Medicine, Department of Physiology, Bilim University, Istanbul, Turkey
| |
Collapse
|
10
|
Neuroprotective effects of Vaccinium myrtillus on damage-related brain injury. J Chem Neuroanat 2023; 127:102193. [PMID: 36414183 DOI: 10.1016/j.jchemneu.2022.102193] [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: 08/28/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Traumatic brain injury may trigger the secondary brain injury, which has the potential to be reversible and thus preventable. Anthocyanins are phylotherapeutic plants, which are reported to exhibit anti-inflammatory properties. This study aimed to evaluate the therapeutic efficiency of an anthocyanin, namely Vaccinium myrtillus, to alleviate secondary brain injury and identify possible mechanism of actions. It is hypothesized that lipid peroxidation and Na+ -K+ -ATPase activity may be involved in neuronal ischemia. Thus, brain tissue Malondialdehyde content, Na+ -K+ -ATPase content, and cleaved caspase-3 content was investigated following moderate head trauma in a rat model. Twenty-four Sprague-Dawley male rats were allocated into four groups: Control, Trauma, Solvent-Control, and Treatment. Trauma and Solvent-Control groups showed more prominent brain edema, neuronal ischemia, vascular congestion, increase in brain tissue Malondialdehyde and cleaved caspase-3 levels, and decreased Na+-K+-ATPase activity compared to the Control group. Although the Treatment group had comparable histological signs to the Trauma and Solvent-Control groups, Malondialdehyde level and Na+-K+-ATPase activity was similar to Control group, and cleaved caspase-3 levels were lower compared to Trauma and Solvent-Control groups. We conclude that anthocyanin extracts may alleviate secondary brain injury via anti-oxidative and anti-apoptotic mechanisms.
Collapse
|
11
|
Nikolaishvili M, Nanobashvili Z, Mitagvaria N, Chkadua G, Museliani T, Jikia G, Bilanishvili I, Dondoladze K. The Level of Individual Biochemical Constants of the Brain of in the Krushinsky-Molodkina Inbred Rat Strain against the Background of Radon Inhalation During Epilepsy. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.10716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Introduction: The elucidation of the mechanism of action of radon on antioxidant processes needs further research, however, based on the results of the experiment, it can be concluded that studies on experimental animals have shown that, while inhaling Tskhaltubo water, the phenomenon of hormesis develops.
Methods: we placed 10 experimental animals (KM rats) in Radon contained mineral water spa`s sauna. Inhalation of radon-water was administered through the nose, for 10 minutes, once a day, in conditions of high humidity (about 90%) for 10 days.
Results: Hormesis regulates oxidative processes in the brain due to the activation of antioxidants expressed in a decrease in existing epileptic seizures and is expressed in the activation of Na / K-ATPase and specific glutaminergic neurons of the "attack center" of the hypothalamus but also with the activation of the entire adaptive-compensatory system.
Conclusion: Inhalation of radon contained water can be considered as a method of treatment with an anticonvulsant effect confirmed by experimental studies.
Collapse
|
12
|
Short-Term Mild Hypoxia Modulates Na,K-ATPase to Maintain Membrane Electrogenesis in Rat Skeletal Muscle. Int J Mol Sci 2022; 23:ijms231911869. [PMID: 36233169 PMCID: PMC9570130 DOI: 10.3390/ijms231911869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
The Na,K-ATPase plays an important role in adaptation to hypoxia. Prolonged hypoxia results in loss of skeletal muscle mass, structure, and performance. However, hypoxic preconditioning is known to protect against a variety of functional impairments. In this study, we tested the possibility of mild hypoxia to modulate the Na,K-ATPase and to improve skeletal muscle electrogenesis. The rats were subjected to simulated high-altitude (3000 m above sea level) hypobaric hypoxia (HH) for 3 h using a hypobaric chamber. Isolated diaphragm and soleus muscles were tested. In the diaphragm muscle, HH increased the α2 Na,K-ATPase isozyme electrogenic activity and stably hyperpolarized the extrajunctional membrane for 24 h. These changes were accompanied by a steady increase in the production of thiobarbituric acid reactive substances as well as a decrease in the serum level of endogenous ouabain, a specific ligand of the Na,K-ATPase. HH also increased the α2 Na,K-ATPase membrane abundance without changing its total protein content; the plasma membrane lipid-ordered phase did not change. In the soleus muscle, HH protected against disuse (hindlimb suspension) induced sarcolemmal depolarization. Considering that the Na,K-ATPase is critical for maintaining skeletal muscle electrogenesis and performance, these findings may have implications for countermeasures in disuse-induced pathology and hypoxic therapy.
Collapse
|
13
|
Chronic Ouabain Prevents Radiation-Induced Reduction in the α2 Na,K-ATPase Function in the Rat Diaphragm Muscle. Int J Mol Sci 2022; 23:ijms231810921. [PMID: 36142836 PMCID: PMC9505176 DOI: 10.3390/ijms231810921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/11/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
The damaging effect of ionizing radiation (IR) on skeletal muscle Na,K-ATPase is an open field of research. Considering a therapeutic potential of ouabain, a specific ligand of the Na,K-ATPase, we tested its ability to protect against the IR-induced disturbances of Na,K-ATPase function in rat diaphragm muscle that co-expresses the α1 and α2 isozymes of this protein. Male Wistar rats (n = 26) were subjected to 6-day injections of vehicle (0.9% NaCl) or ouabain (1 µg/kg/day). On the fourth day of injections, rats were exposed to one-time total-body X-ray irradiation (10 Gy), or a sham irradiation. The isolated muscles were studied 72 h post-irradiation. IR decreased the electrogenic contribution of the α2 Na,K-ATPase without affecting its protein content, thereby causing sarcolemma depolarization. IR increased serum concentrations of ouabain, IL-6, and corticosterone, decreased lipid peroxidation, and changed cellular redox status. Chronic ouabain administration prevented IR-induced depolarization and loss of the α2 Na,K-ATPase electrogenic contribution without changing its protein content. This was accompanied with an elevation of ouabain concentration in circulation and with the lack of IR-induced suppression of lipid peroxidation. Given the crucial role of Na,K-ATPase in skeletal muscle performance, these findings may have therapeutic implications as countermeasures for IR-induced muscle pathology.
Collapse
|
14
|
Xu Y, Fan Q. Relationship between chronic hypoxia and seizure susceptibility. CNS Neurosci Ther 2022; 28:1689-1705. [PMID: 35983626 PMCID: PMC9532927 DOI: 10.1111/cns.13942] [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: 10/21/2021] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/16/2023] Open
Abstract
Chronic hypobaric hypoxia in high‐altitude areas is closely related to the occurrence of many neurological diseases. Among these diseases, epilepsy is a common disease of the nervous system that is difficult to diagnose and treat, with a long treatment cycle. As of 2019, there were more than 70 million epilepsy patients worldwide, including 10 million in China. Studies have shown that chronic hypoxia promotes the occurrence and development of epilepsy, and elucidation of the relationship between chronic hypoxia and epilepsy is important for studying the pathogenesis of epilepsy and exploring the potential characteristics of epilepsy and new drug targets for epilepsy. In this article, we review the factors that may cause increased seizure susceptibility in chronic hypoxia and consider the potential relationship between chronic hypobaric hypoxia and seizure susceptibility in high‐altitude areas and prospects surrounding related research in the future.
Collapse
Affiliation(s)
- YuanHang Xu
- Qinghai University Graduate School, Xining, China.,Department of Neurology, Qinghai Provincial People's Hospital Xining, Xining, China
| | - QingLi Fan
- Department of Neurology, Qinghai Provincial People's Hospital Xining, Xining, China
| |
Collapse
|
15
|
Andrade P, Lara-Valderrábano L, Manninen E, Ciszek R, Tapiala J, Ndode-Ekane XE, Pitkänen A. Seizure Susceptibility and Sleep Disturbance as Biomarkers of Epileptogenesis after Experimental TBI. Biomedicines 2022; 10:biomedicines10051138. [PMID: 35625875 PMCID: PMC9138230 DOI: 10.3390/biomedicines10051138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
Objectives: We investigated whether seizure susceptibility increases over weeks−months after experimental traumatic brain injury (TBI), and whether seizure susceptibility in rats predicts the development of post-traumatic epilepsy (PTE) or epileptiform activity. We further investigated whether rats develop chronic sleep disturbance after TBI, and whether sleep disturbance parameters—alone or in combination with pentylenetetrazol (PTZ) test parameters—could serve as novel biomarkers for the development of post-traumatic epileptogenesis. Methods: TBI was induced in adult male Sprague-Dawley rats with lateral fluid-percussion injury. Sham-operated experimental controls underwent craniectomy without exposure to an impact force. Seizure susceptibility was tested with a PTZ test (30 mg/kg, intraperitoneally) on day (D) 30, D60, D90, and D180 after TBI (n = 28) or sham operation (n = 16) under video electroencephalogram (vEEG). In the 7th post-injury month, rats underwent continuous vEEG monitoring to detect spontaneous seizures and assess sleep disturbances. At the end of the experiments, rats were perfused for brain histology. Results: In the TBI group, the percentage of rats with PTZ-induced seizures increased over time (adjusted p < 0.05 compared with D30). Combinations of three PTZ test parameters (latency to the first epileptiform discharge (ED), number of EDs, and number of PTZ-induced seizures) survived the leave-one-out validation for differentiating rats with or without epileptiform activity, indicating an area under the receiver operating curve (AUC) of 0.743 (95% CI 0.472−0.992, p = 0.05) with a misclassification rate of 36% on D90, and an AUC of 0.752 (95% CI 0.483−0.929, p < 0.05) with a misclassification rate of 32% on D180. Sleep analysis revealed that the number of transitions to N3 or rapid eye movement (REM) sleep, along with the total number of transitions, was increased in the TBI group during the lights-on period (all p < 0.05). The sleep fragmentation index during the lights-on period was greater in the TBI rats than in sham-operated rats (p < 0.05). A combination of sleep parameters showed promise as diagnostic biomarkers of prior TBI, with an AUC of 0.792 (95% CI 0.549−0.934, p < 0.01) and a misclassification rate of 28%. Rats with epilepsy or any epileptiform activity had more transitions from N3 to the awake stage (p < 0.05), and the number of N3−awake transitions differentiated rats with or without epileptiform activity, with an AUC of 0.857 (95% CI 0.651−1.063, p < 0.01). Combining sleep parameters with PTZ parameters did not improve the biomarker performance. Significance: This is the first attempt to monitor the evolution of seizure susceptibility over months in a well-described rat model of PTE. Our data suggest that assessment of seizure susceptibility and sleep disturbance can provide diagnostic biomarkers of prior TBI and prognostic biomarkers of post-traumatic epileptogenesis.
Collapse
|
16
|
Selected Molecular Targets for Antiepileptogenesis. Int J Mol Sci 2021; 22:ijms22189737. [PMID: 34575901 PMCID: PMC8466306 DOI: 10.3390/ijms22189737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023] Open
Abstract
The term epileptogenesis defines the usually durable process of converting normal brain into an epileptic one. The resistance of a significant proportion of patients with epilepsy to the available pharmacotherapy prompted the concept of a causative treatment option consisting in stopping or modifying the progress of epileptogenesis. Most antiepileptic drugs possess only a weak or no antiepileptogenic potential at all, but a few of them appear promising in this regard; these include, for example, eslicarbazepine (a sodium and T-type channel blocker), lamotrigine (a sodium channel blocker and glutamate antagonist) or levetiracetam (a ligand of synaptic vehicle protein SV2A). Among the approved non-antiepileptic drugs, antiepileptogenic potential seems to reside in losartan (a blocker of angiotensin II type 1 receptors), biperiden (an antiparkinsonian drug), nonsteroidal anti-inflammatory drugs, antioxidative drugs and minocycline (a second-generation tetracycline with anti-inflammatory and antioxidant properties). Among other possible antiepileptogenic compounds, antisense nucleotides have been considered, among these an antagomir targeting microRNA-134. The drugs and agents mentioned above have been evaluated in post-status epilepticus models of epileptogenesis, so their preventive efficacy must be verified. Limited clinical data indicate that biperiden in patients with brain injuries is well-tolerated and seems to reduce the incidence of post-traumatic epilepsy. Exceptionally, in this regard, our own original data presented here point to c-Fos as an early seizure duration, but not seizure intensity-related, marker of early epileptogenesis. Further research of reliable markers of early epileptogenesis is definitely needed to improve the process of designing adequate antiepileptogenic therapies.
Collapse
|
17
|
Fourier Transform Infrared Imaging-A Novel Approach to Monitor Bio Molecular Changes in Subacute Mild Traumatic Brain Injury. Brain Sci 2021; 11:brainsci11070918. [PMID: 34356152 PMCID: PMC8307811 DOI: 10.3390/brainsci11070918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) can be defined as a disorder in the function of the brain after a bump, blow, or jolt to the head, or penetrating head injury. Mild traumatic brain injury (mTBI) can cause devastating effects, such as the initiation of long-term neurodegeneration in brain tissue. In the current study, the effects of mTBI were investigated on rat brain regions; cortex (Co) and corpus callosum (CC) after 24 h (subacute trauma) by Fourier transform infrared (FTIR) imaging and immunohistochemistry (IHC). IHC studies showed the formation of amyloid-β (Aβ) plaques in the cortex brain region of mTBI rats. Moreover, staining of myelin basic protein presented the shearing of axons in CC region in the same group of animals. According to FTIR imaging results, total protein and lipid content significantly decreased in both Co and CC regions in mTBI group compared to the control. Due to this significant decrease in both lipid and protein content, remarkable consistency in lipid/protein band ratio in mTBI and control group, was observed. Significant decrease in methyl content and a significant increase in olefinic content were observed in Co and CC regions of mTBI rat brain tissues. Classification amongst distinguishable groups was performed using principal component analysis (PCA) and hierarchical clustering (HCA). This study established the prospective of FTIR imaging for assessing biochemical changes due to mTBI with high sensitivity, precision and high-resolution.
Collapse
|
18
|
Vigil FA, Bozdemir E, Bugay V, Chun SH, Hobbs M, Sanchez I, Hastings SD, Veraza RJ, Holstein DM, Sprague SM, M Carver C, Cavazos JE, Brenner R, Lechleiter JD, Shapiro MS. Prevention of brain damage after traumatic brain injury by pharmacological enhancement of KCNQ (Kv7, "M-type") K + currents in neurons. J Cereb Blood Flow Metab 2020; 40:1256-1273. [PMID: 31272312 PMCID: PMC7238379 DOI: 10.1177/0271678x19857818] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nearly three million people in the USA suffer traumatic brain injury (TBI) yearly; however, there are no pre- or post-TBI treatment options available. KCNQ2-5 voltage-gated K+ channels underlie the neuronal "M current", which plays a dominant role in the regulation of neuronal excitability. Our strategy towards prevention of TBI-induced brain damage is predicated on the suggested hyper-excitability of neurons induced by TBIs, and the decrease in neuronal excitation upon pharmacological augmentation of M/KCNQ K+ currents. Seizures are very common after a TBI, making further seizures and development of epilepsy disease more likely. Our hypothesis is that TBI-induced hyperexcitability and ischemia/hypoxia lead to metabolic stress, cell death and a maladaptive inflammatory response that causes further downstream morbidity. Using the mouse controlled closed-cortical impact blunt TBI model, we found that systemic administration of the prototype M-channel "opener", retigabine (RTG), 30 min after TBI, reduces the post-TBI cascade of events, including spontaneous seizures, enhanced susceptibility to chemo-convulsants, metabolic stress, inflammatory responses, blood-brain barrier breakdown, and cell death. This work suggests that acutely reducing neuronal excitability and energy demand via M-current enhancement may be a novel model of therapeutic intervention against post-TBI brain damage and dysfunction.
Collapse
Affiliation(s)
- Fabio A Vigil
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Vladislav Bugay
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - MaryAnn Hobbs
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Isamar Sanchez
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shayne D Hastings
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Rafael J Veraza
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shane M Sprague
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jose E Cavazos
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Robert Brenner
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mark S Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
19
|
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: 62] [Impact Index Per Article: 12.4] [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
|
20
|
Eslami M, Alizadeh L, Morteza-Zadeh P, Sayyah M. The effect of Lipopolysaccharide (LPS) pretreatment on hippocampal apoptosis in traumatic rats. Neurol Res 2020; 42:91-98. [PMID: 31914902 DOI: 10.1080/01616412.2019.1709139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: Traumatic brain injury (TBI) is a serious medical problem that affects the quality of life. Apoptosis is a form of programmed cell death that happens after trauma. Effector caspases are responsible for initiating apoptosis.Methods: In the present study, we examined the effect of LPS preconditioning (0.1 and 0.5 mg/kg, ip; 5 days prior controlled cortical injury) on apoptosis, 4 and 12 hours after trauma. We investigated possible mechanisms on the expression of caspase3 and caspase7 in hippocampal CA1 and CA3 areas by using immunohistochemistry and Western blotting techniques and also TUNEL-positive cells.Results: Higher expression of caspase3 and caspase7 were accompanied by a higher number of dead neurons in traumatic rats 4 and 12 hours after trauma(P < 0.05). LPS preconditioning decreased caspase3 and caspase7over-expression and the number of dead neurons in the hippocampus(P < 0.05).Discussion: Our data indicate that LPS preconditioning inhibits neural damage and apoptosis induced by trauma in the hippocampus.
Collapse
Affiliation(s)
- Mansoureh Eslami
- Department of Basic Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Alizadeh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Sousa L, Oliveira MM, Pessôa MTC, Barbosa LA. Iron overload: Effects on cellular biochemistry. Clin Chim Acta 2019; 504:180-189. [PMID: 31790701 DOI: 10.1016/j.cca.2019.11.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
Iron is an essential element for human life. However, it is a pro-oxidant agent capable of reacting with hydrogen peroxide. An iron overload can cause cellular changes, such as damage to the plasma membrane leading to cell death. Effects of iron overload in cellular biochemical processes include modulating membrane enzymes, such as the Na, K-ATPase, impairing the ionic transport and inducing irreversible damage to cellular homeostasis. To avoid such damage, cells have an antioxidant system that acts in an integrated manner to prevent oxidative stress. In addition, the cells contain proteins responsible for iron transport and storage, preventing its reaction with other substances during absorption. Moreover, iron is associated with cellular events coordinated by iron-responsive proteins (IRPs) that regulate several cellular functions, including a process of cell death called ferroptosis. This review will address the biochemical aspects of iron overload at the cellular level and its effects on important cellular structures.
Collapse
Affiliation(s)
- Leilismara Sousa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Marina M Oliveira
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Marco Túlio C Pessôa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Leandro A Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil.
| |
Collapse
|
22
|
Terrone G, Balosso S, Pauletti A, Ravizza T, Vezzani A. Inflammation and reactive oxygen species as disease modifiers in epilepsy. Neuropharmacology 2019; 167:107742. [PMID: 31421074 DOI: 10.1016/j.neuropharm.2019.107742] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/10/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation and reactive oxygen and nitrogen species are rapidly induced in the brain after acute cerebral injuries that are associated with an enhanced risk for epilepsy in humans and related animal models. These phenomena reinforce each others and persist during epileptogenesis as well as during chronic spontaneous seizures. Anti-inflammatory and anti-oxidant drugs transiently administered either before, or shortly after the clinical onset of symptomatic epilepsy, similarly block the progression of spontaneous seizures, and may delay their onset. Moreover, neuroprotection and rescue of cognitive deficits are also observed in the treated animals. Therefore, although these treatments do not prevent epilepsy development, they offer clinically relevant disease-modification effects. These therapeutic effects are mediated by targeting molecular signaling pathways such as the IL-1β-IL-1 receptor type 1 and TLR4, P2X7 receptors, the transcriptional anti-oxidant factor Nrf2, while the therapeutic impact of COX-2 inhibition for reducing spontaneous seizures remains controversial. Some anti-inflammatory and anti-oxidant drugs that are endowed of disease modification effects in preclinical models are already in medical use and have a safety profile, therefore, they provide potential re-purposed treatments for improving the disease course and for reducing seizure burden. Markers of neuroinflammation and oxidative stress can be measured in blood or by neuroimaging, therefore they represent testable prognostic and predictive biomarkers for selecting the patient's population at high risk for developing epilepsy therefore eligible for novel treatments. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Gaetano Terrone
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Silvia Balosso
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Alberto Pauletti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Teresa Ravizza
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Annamaria Vezzani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| |
Collapse
|
23
|
Pauletti A, Terrone G, Shekh-Ahmad T, Salamone A, Ravizza T, Rizzi M, Pastore A, Pascente R, Liang LP, Villa BR, Balosso S, Abramov AY, van Vliet EA, Del Giudice E, Aronica E, Patel M, Walker MC, Vezzani A. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain 2019; 142:e39. [PMID: 31145451 PMCID: PMC6598637 DOI: 10.1093/brain/awz130] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/17/2017] [Accepted: 03/26/2017] [Indexed: 01/07/2023] Open
Abstract
Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.
Collapse
Affiliation(s)
- Alberto Pauletti
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Gaetano Terrone
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Tawfeeq Shekh-Ahmad
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Alessia Salamone
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Teresa Ravizza
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Massimo Rizzi
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Anna Pastore
- 3 Metabolomics and Proteomics Unit, ‘Bambino Gesù’ Children’s Hospital,
IRCCS, Rome, Italy
| | - Rosaria Pascente
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Li-Ping Liang
- 4 Department of Pharmaceutical Sciences, University of Colorado Denver,
Aurora, Colorado, USA
| | - Bianca R Villa
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Silvia Balosso
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
| | - Andrey Y Abramov
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Erwin A van Vliet
- 5 Department of (Neuro)Pathology, Academic Medical Center, University of
Amsterdam, The Netherlands
| | - Ennio Del Giudice
- 6 Department of Translational Medical Sciences, Section of Pediatrics,
Federico II University, Naples, Italy
| | - Eleonora Aronica
- 5 Department of (Neuro)Pathology, Academic Medical Center, University of
Amsterdam, The Netherlands
- 7 Stichting Epilepsie Instellingen Nederland, Amsterdam, The
Netherlands
| | - Manisha Patel
- 4 Department of Pharmaceutical Sciences, University of Colorado Denver,
Aurora, Colorado, USA
| | - Matthew C Walker
- 2 Department of Clinical and Experimental Epilepsy, University College
London, UK
| | - Annamaria Vezzani
- 1 Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche
Mario Negri, Milan, Italy
- Correpondence to: Annamaria Vezzani, PhD Department of Neuroscience
IRCCS-Istituto di Ricerche Farmacologiche Mario Negri Via G. La Masa 19, 20156 Milano,
Italy E-mail:
| |
Collapse
|
24
|
Black berry juice attenuates neurological disorders and oxidative stress associated with concurrent exposure of aluminum and fluoride in male rats. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.ejbas.2015.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Ravizza T, Vezzani A. Pharmacological targeting of brain inflammation in epilepsy: Therapeutic perspectives from experimental and clinical studies. Epilepsia Open 2018; 3:133-142. [PMID: 30564772 PMCID: PMC6293065 DOI: 10.1002/epi4.12242] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2018] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence supports a pathogenic role of unabated neuroinflammation in various central nervous system (CNS) diseases, including epilepsy. Neuroinflammation is not a bystander phenomenon of the diseased brain tissue, but it may contribute to neuronal hyperexcitability underlying seizure generation, cell loss, and neurologic comorbidities. Several molecules, which constitute the inflammatory milieu in the epileptogenic area, activate signaling pathways in neurons and glia resulting in pathologic modifications of cell function, which ultimately lead to alterations in synaptic transmission and plasticity. Herein we report the up-to-date experimental and clinical evidence that supports the neuromodulatory role of inflammatory mediators, their related signaling pathways, and involvement in epilepsy. We discuss how these mechanisms can be harnessed to discover and validate targets for novel therapeutics, which may prevent or control pharmacoresistant epilepsies.
Collapse
Affiliation(s)
- Teresa Ravizza
- Department of NeuroscienceIRCCS – Mario Negri Institute for Pharmacological ResearchMilanoItaly
| | - Annamaria Vezzani
- Department of NeuroscienceIRCCS – Mario Negri Institute for Pharmacological ResearchMilanoItaly
| |
Collapse
|
26
|
Arulsamy A, Teng J, Colton H, Corrigan F, Collins-Praino L. Evaluation of early chronic functional outcomes and their relationship to pre-frontal cortex and hippocampal pathology following moderate-severe traumatic brain injury. Behav Brain Res 2018; 348:127-138. [DOI: 10.1016/j.bbr.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
|
27
|
Bhatti J, Nascimento B, Akhtar U, Rhind SG, Tien H, Nathens A, da Luz LT. Systematic Review of Human and Animal Studies Examining the Efficacy and Safety of N-Acetylcysteine (NAC) and N-Acetylcysteine Amide (NACA) in Traumatic Brain Injury: Impact on Neurofunctional Outcome and Biomarkers of Oxidative Stress and Inflammation. Front Neurol 2018; 8:744. [PMID: 29387038 PMCID: PMC5776005 DOI: 10.3389/fneur.2017.00744] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Background No new therapies for traumatic brain injury (TBI) have been officially translated into current practice. At the tissue and cellular level, both inflammatory and oxidative processes may be exacerbated post-injury and contribute to further brain damage. N-acetylcysteine (NAC) has the potential to downregulate both processes. This review focuses on the potential neuroprotective utility of NAC and N-acetylcysteine amide (NACA) post-TBI. Methods Medline, Embase, Cochrane Library, and ClinicalTrials.gov were searched up to July 2017. Studies that examined clinical and laboratory effects of NAC and NACA post-TBI in human and animal studies were included. Risk of bias was assessed in human and animal studies according to the design of each study (randomized or not). The primary outcome assessed was the effect of NAC/NACA treatment on functional outcome, while secondary outcomes included the impact on biomarkers of inflammation and oxidation. Due to the clinical and methodological heterogeneity observed across studies, no meta-analyses were conducted. Results Our analyses revealed only three human trials, including two randomized controlled trials (RCTs) and 20 animal studies conducted using standardized animal models of brain injury. The two RCTs reported improvement in the functional outcome post-NAC/NACA administration. Overall, the evidence from animal studies is more robust and demonstrated substantial improvement of cognition and psychomotor performance following NAC/NACA use. Animal studies also reported significantly more cortical sparing, reduced apoptosis, and lower levels of biomarkers of inflammation and oxidative stress. No safety concerns were reported in any of the studies included in this analysis. Conclusion Evidence from the animal literature demonstrates a robust association for the prophylactic application of NAC and NACA post-TBI with improved neurofunctional outcomes and downregulation of inflammatory and oxidative stress markers at the tissue level. While a growing body of scientific literature suggests putative beneficial effects of NAC/NACA treatment for TBI, the lack of well-designed and controlled clinical investigations, evaluating therapeutic outcomes, prognostic biomarkers, and safety profiles, limits definitive interpretation and recommendations for its application in humans at this time.
Collapse
Affiliation(s)
- Junaid Bhatti
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Barto Nascimento
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Umbreen Akhtar
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Shawn G Rhind
- Defense Research and Development Canada (DRDC), Toronto Research Centre, Toronto, ON, Canada
| | - Homer Tien
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Avery Nathens
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Luis Teodoro da Luz
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Chandel S, Gupta SK, Medhi B. Epileptogenesis following experimentally induced traumatic brain injury - a systematic review. Rev Neurosci 2018; 27:329-46. [PMID: 26581067 DOI: 10.1515/revneuro-2015-0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a complex neurotrauma in civilian life and the battlefield with a broad spectrum of symptoms, long-term neuropsychological disability, as well as mortality worldwide. Posttraumatic epilepsy (PTE) is a common outcome of TBI with unknown mechanisms, followed by posttraumatic epileptogenesis. There are numerous rodent models of TBI available with varying pathomechanisms of head injury similar to human TBI, but there is no evidence for an adequate TBI model that can properly mimic all aspects of clinical TBI and the first successive spontaneous focal seizures follow a single episode of neurotrauma with respect to epileptogenesis. This review aims to provide current information regarding the various experimental animal models of TBI relevant to clinical TBI. Mossy fiber sprouting, loss of dentate hilar neurons along with recurrent seizures, and epileptic discharge similar to human PTE have been studied in fluid percussion injury, weight-drop injury, and cortical impact models, but further refinement of animal models and functional test is warranted to better understand the underlying pathophysiology of posttraumatic epileptogenesis. A multifaceted research approach in TBI model may lead to exploration of the potential treatment measures, which are a major challenge to the research community and drug developers. With respect to clinical setting, proper patient data collection, improved clinical trials with advancement in drug delivery strategies, blood-brain barrier permeability, and proper monitoring of level and effects of target drug are also important.
Collapse
|
29
|
Parreira GM, Resende MDA, Garcia IJP, Sartori DB, Umeoka EHDL, Godoy LD, Garcia-Cairasco N, Barbosa LA, Santos HDL, Tilelli CQ. Oxidative stress and Na,K-ATPase activity differential regulation in brainstem and forebrain of Wistar Audiogenic rats may lead to increased seizure susceptibility. Brain Res 2018; 1679:171-178. [DOI: 10.1016/j.brainres.2017.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 11/16/2022]
|
30
|
Bazzigaluppi P, Ebrahim Amini A, Weisspapir I, Stefanovic B, Carlen PL. Hungry Neurons: Metabolic Insights on Seizure Dynamics. Int J Mol Sci 2017; 18:ijms18112269. [PMID: 29143800 PMCID: PMC5713239 DOI: 10.3390/ijms18112269] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
Epilepsy afflicts up to 1.6% of the population and the mechanisms underlying the appearance of seizures are still not understood. In past years, many efforts have been spent trying to understand the mechanisms underlying the excessive and synchronous firing of neurons. Traditionally, attention was pointed towards synaptic (dys)function and extracellular ionic species (dys)regulation. Recently, novel clinical and preclinical studies explored the role of brain metabolism (i.e., glucose utilization) of seizures pathophysiology revealing (in most cases) reduced metabolism in the inter-ictal period and increased metabolism in the seconds preceding and during the appearance of seizures. In the present review, we summarize the clinical and preclinical observations showing metabolic dysregulation during epileptogenesis, seizure initiation, and termination, and in the inter-ictal period. Recent preclinical studies have shown that 2-Deoxyglucose (2-DG, a glycolysis blocker) is a novel therapeutic approach to reduce seizures. Furthermore, we present initial evidence for the effectiveness of 2-DG in arresting 4-Aminopyridine induced neocortical seizures in vivo in the mouse.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- Krembil Research Institute, Fundamental Neurobiology, Toronto, ON M5T 2S8, Canada.
- Sunnybrook Research Institute, Medical Biophysics, Toronto, ON M4N 3M5, Canada.
| | - Azin Ebrahim Amini
- Krembil Research Institute, Fundamental Neurobiology, Toronto, ON M5T 2S8, Canada.
- Institute of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, ON M5S 3G9, Canada.
| | - Iliya Weisspapir
- Krembil Research Institute, Fundamental Neurobiology, Toronto, ON M5T 2S8, Canada.
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Medical Biophysics, Toronto, ON M4N 3M5, Canada.
| | - Peter L Carlen
- Krembil Research Institute, Fundamental Neurobiology, Toronto, ON M5T 2S8, Canada.
- Department of Medicine & Physiology, and Institute of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
31
|
Pauletti A, Terrone G, Shekh-Ahmad T, Salamone A, Ravizza T, Rizzi M, Pastore A, Pascente R, Liang LP, Villa BR, Balosso S, Abramov AY, van Vliet EA, Del Giudice E, Aronica E, Antoine DJ, Patel M, Walker MC, Vezzani A. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain 2017; 140:1885-1899. [PMID: 28575153 DOI: 10.1093/brain/awx117] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/26/2017] [Indexed: 12/31/2022] Open
Abstract
Epilepsy therapy is based on antiseizure drugs that treat the symptom, seizures, rather than the disease and are ineffective in up to 30% of patients. There are no treatments for modifying the disease-preventing seizure onset, reducing severity or improving prognosis. Among the potential molecular targets for attaining these unmet therapeutic needs, we focused on oxidative stress since it is a pathophysiological process commonly occurring in experimental epileptogenesis and observed in human epilepsy. Using a rat model of acquired epilepsy induced by electrical status epilepticus, we show that oxidative stress occurs in both neurons and astrocytes during epileptogenesis, as assessed by measuring biochemical and histological markers. This evidence was validated in the hippocampus of humans who died following status epilepticus. Oxidative stress was reduced in animals undergoing epileptogenesis by a transient treatment with N-acetylcysteine and sulforaphane, which act to increase glutathione levels through complementary mechanisms. These antioxidant drugs are already used in humans for other therapeutic indications. This drug combination transiently administered for 2 weeks during epileptogenesis inhibited oxidative stress more efficiently than either drug alone. The drug combination significantly delayed the onset of epilepsy, blocked disease progression between 2 and 5 months post-status epilepticus and drastically reduced the frequency of spontaneous seizures measured at 5 months without modifying the average seizure duration or the incidence of epilepsy in animals. Treatment also decreased hippocampal neuron loss and rescued cognitive deficits. Oxidative stress during epileptogenesis was associated with de novo brain and blood generation of disulfide high mobility group box 1 (HMGB1), a neuroinflammatory molecule implicated in seizure mechanisms. Drug-induced reduction of oxidative stress prevented disulfide HMGB1 generation, thus highlighting a potential novel mechanism contributing to therapeutic effects. Our data show that targeting oxidative stress with clinically used drugs for a limited time window starting early after injury significantly improves long-term disease outcomes. This intervention may be considered for patients exposed to potential epileptogenic insults.
Collapse
Affiliation(s)
- Alberto Pauletti
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Gaetano Terrone
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Tawfeeq Shekh-Ahmad
- Department of Clinical and Experimental Epilepsy, University College London, UK
| | - Alessia Salamone
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Teresa Ravizza
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Massimo Rizzi
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Anna Pastore
- Metabolomics and Proteomics Unit, 'Bambino Gesù' Children's Hospital, IRCCS, Rome, Italy
| | - Rosaria Pascente
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Bianca R Villa
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Silvia Balosso
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Andrey Y Abramov
- Department of Clinical and Experimental Epilepsy, University College London, UK
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Ennio Del Giudice
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University, Naples, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland, Amsterdam, The Netherlands
| | - Daniel J Antoine
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, University College London, UK
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
32
|
Wang CQ, Ye Y, Chen F, Han WC, Sun JM, Lu X, Guo R, Cao K, Zheng MJ, Liao LC. Posttraumatic administration of a sub-anesthetic dose of ketamine exerts neuroprotection via attenuating inflammation and autophagy. Neuroscience 2016; 343:30-38. [PMID: 27916727 DOI: 10.1016/j.neuroscience.2016.11.029] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 02/05/2023]
Abstract
As a complex disease, traumatic brain injury (TBI) can result in long-term psychiatric changes and sensorimotor and cognitive impairments. The TBI-induced loss of memory and long-term cognitive dysfunction are related to mechanistic factors including an increased inflammatory response, autophagy, edema, and ischemia. Many published studies have offered evidence for the neuroprotective effects and anti-inflammatory properties of ketamine for TBI patients. Nonetheless, there is a limited understanding of the accurate mechanism that underlies the potential neuroprotective effects of ketamine. Herein, it can be shown that posttraumatic administration of ketamine at a sub-anesthetic dose (10mg/kg ketamine, every 24h up to 7days) can prevent the TBI-induced production of IL-6 and TNF-α, attenuate deficits of dendrites and spines and exert beneficial effects on memory and behavior. Moreover, studies show that ketamine may activate the mTOR signaling pathway by p-mTOR induction to down-regulate the expression of crucial autophagic proteins such as LC3 and Beclin-1. According to these findings, ameliorating secondary brain injury and anti-inflammatory properties is closely related to the neuroprotection of ketamine, which supports the use of ketamine as a potential therapy for patients with TBI to alleviate functional deficits.
Collapse
Affiliation(s)
- C-Q Wang
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China; Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - Y Ye
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - F Chen
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - W-C Han
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - J-M Sun
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - X Lu
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - R Guo
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - K Cao
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - M-J Zheng
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - L-C Liao
- College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
33
|
Adefegha SA, Oboh G, Omojokun OS, Adefegha OM. Alterations of Na+/K+-ATPase, cholinergic and antioxidant enzymes activity by protocatechuic acid in cadmium-induced neurotoxicity and oxidative stress in Wistar rats. Biomed Pharmacother 2016; 83:559-568. [DOI: 10.1016/j.biopha.2016.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 12/21/2022] Open
|
34
|
Das J, Singh R, Sharma D. Antiepileptic effect of fisetin in iron-induced experimental model of traumatic epilepsy in rats in the light of electrophysiological, biochemical, and behavioral observations. Nutr Neurosci 2016; 20:255-264. [DOI: 10.1080/1028415x.2016.1183342] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jharana Das
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rameshwar Singh
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Deepak Sharma
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
35
|
Yonutas HM, Vekaria HJ, Sullivan PG. Mitochondrial specific therapeutic targets following brain injury. Brain Res 2016; 1640:77-93. [PMID: 26872596 DOI: 10.1016/j.brainres.2016.02.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 02/03/2023]
Abstract
Traumatic brain injury is a complicated disease to treat due to the complex multi-factorial secondary injury cascade that is initiated following the initial impact. This secondary injury cascade causes nonmechanical tissue damage, which is where therapeutic interventions may be efficacious for intervention. One therapeutic target that has shown much promise following brain injury are mitochondria. Mitochondria are complex organelles found within the cell. At a superficial level, mitochondria are known to produce the energy substrate used within the cell called ATP. However, their importance to overall cellular homeostasis is even larger than their production of ATP. These organelles are necessary for calcium cycling, ROS production and play a role in the initiation of cell death pathways. When mitochondria become dysfunctional, they can become dysregulated leading to a loss of cellular homeostasis and eventual cell death. Within this review there will be a deep discussion into mitochondrial bioenergetics followed by a brief discussion into traumatic brain injury and how mitochondria play an integral role in the neuropathological sequelae following an injury. The review will conclude with a discussion pertaining to the therapeutic approaches currently being studied to ameliorate mitochondrial dysfunction following brain injury. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
Affiliation(s)
- H M Yonutas
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States
| | - H J Vekaria
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States
| | - P G Sullivan
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States.
| |
Collapse
|
36
|
Poersch AB, Trombetta F, Souto NS, de Oliveira Lima C, Braga ACM, Dobrachinski F, Ribeiro LR, Soares FAA, Fighera MR, Royes LFF, Oliveira MS, Furian AF. Fumonisin B1 facilitates seizures induced by pentylenetetrazol in mice. Neurotoxicol Teratol 2015; 51:61-7. [PMID: 26342287 DOI: 10.1016/j.ntt.2015.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
Abstract
Fumonisin B1 (FB1) is a Fusarium spp. mycotoxin which constitutes a major public health issue because of its worldwide distribution and diversity of toxic effects.While the liver and kidney are considered the major target organs of FB1 toxicity in several species, evidence indicates that FB1 may be toxic to the brain. To further investigate the effects of FB1 on the central nervous system the present study aimed to test the hypothesis that acute FB1 exposure causes brain hyperexcitability and the potential underlying mechanisms. For these purposes, adult male C57BL/6 mice were injected with FB1 (8 mg/kg, i.p.) or its vehicle and 30 min thereafter received with a low dose of the classical convulsant pentylenetetrazol (PTZ, 30 mg/kg, i.p.) or its vehicle. After behavioral evaluation the cerebral cortex and the hippocampus were collected for analysis of Na(+),K(+)-ATPase activity, mitochondrial membrane potential (ΔΨm) and mitochondrial complex I and II activities. We found that FB1 reduced the latency for PTZ-induced myoclonic jerks and increased the number of these events. After exposure to FB1 total and α1 Na(+),K(+)-ATPase activities increased in cerebral cortex, whereas the same enzyme activities decreased in the hippocampus. Although no changes in mitochondrial complex I and II activities were found, acute exposure to FB1 increased ΔΨm in the cerebral cortex. Altogether, present results indicate that FB1 causes brain hyperexcitability in vivo, and that mitochondrial dysfunction may represent a potential underlying mechanism.
Collapse
Affiliation(s)
- Alice Bertotto Poersch
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Francielle Trombetta
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Naiéli Schiefelbein Souto
- Programa de Pós Graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Camilla de Oliveira Lima
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Ana Cláudia Monteiro Braga
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Fernando Dobrachinski
- Programa de Pós Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Campus UFSM, Santa Maria, RS 97105-900, Brazil
| | - Leandro Rodrigo Ribeiro
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Félix Alexandre Antunes Soares
- Programa de Pós Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Campus UFSM, Santa Maria, RS 97105-900, Brazil
| | - Michele Rechia Fighera
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Luiz Fernando Freire Royes
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil; Programa de Pós Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Campus UFSM, Santa Maria, RS 97105-900, Brazil
| | - Mauro Schneider Oliveira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil; Programa de Pós Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Campus UFSM, Santa Maria, RS 97105-900, Brazil
| | - Ana Flávia Furian
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil; Programa de Pós Graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
37
|
Carvalho FB, Gutierres JM, Bohnert C, Zago AM, Abdalla FH, Vieira JM, Palma HE, Oliveira SM, Spanevello RM, Duarte MM, Lopes ST, Aiello G, Amaral MG, Pippi NL, Andrade CM. Anthocyanins suppress the secretion of proinflammatory mediators and oxidative stress, and restore ion pump activities in demyelination. J Nutr Biochem 2015; 26:378-90. [DOI: 10.1016/j.jnutbio.2014.11.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 10/09/2014] [Accepted: 11/13/2014] [Indexed: 12/23/2022]
|
38
|
Pires LF, Costa LM, de Almeida AAC, Silva OA, Cerqueira GS, de Sousa DP, Pires RMC, Satyal P, de Freitas RM. Neuropharmacological effects of carvacryl acetate on δ-aminolevulinic dehydratase, Na+, K+-ATPase activities and amino acids levels in mice hippocampus after seizures. Chem Biol Interact 2015; 226:49-57. [DOI: 10.1016/j.cbi.2014.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 12/23/2022]
|
39
|
Martinc B, Grabnar I, Vovk T. Antioxidants as a preventive treatment for epileptic process: a review of the current status. Curr Neuropharmacol 2014; 12:527-50. [PMID: 25977679 PMCID: PMC4428026 DOI: 10.2174/1570159x12666140923205715] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/29/2014] [Accepted: 09/22/2014] [Indexed: 11/25/2022] Open
Abstract
Epilepsy is known as one of the most frequent neurological diseases, characterized by an enduring predisposition to generate epileptic seizures. Oxidative stress is believed to directly participate in pathways leading to neurodegeneration, which serves as the most important propagating factor, leading to the epileptic condition and cognitive decline. Moreover, there is also a growing body of evidence showing the disturbance of antioxidant system balance and consequently increased production of reactive species in patients with epilepsy. A meta-analysis, conducted in the present review confirms an association between epilepsy and increased lipid peroxidation. Furthermore, it was also shown that some of the antiepileptic drugs could potentially be responsible for additionally increased lipid peroxidation. Therefore, it is reasonable to propose that during the epileptic process neuroprotective treatment with antioxidants could lead to less sever structural damages, reduced epileptogenesis and milder cognitive deterioration. To evaluate this hypothesis studies investigating the neuroprotective therapeutic potential of various antioxidants in cells, animal seizure models and patients with epilepsy have been reviewed. Numerous beneficial effects of antioxidants on oxidative stress markers and in some cases also neuroprotective effects were observed in animal seizure models. However, despite these encouraging results, till now only a few antioxidants have been further applied to patients with epilepsy as an add-on therapy. Based on the several positive findings in animal models, a strong need for more carefully planned, randomized, double-blind, cross-over, placebo-controlled clinical trials for the evaluation of antioxidants efficacy in patients with epilepsy is warranted.
Collapse
Affiliation(s)
| | | | - Tomaž Vovk
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
40
|
Imbrosci B, Neitz A, Mittmann T. Focal cortical lesions induce bidirectional changes in the excitability of fast spiking and non fast spiking cortical interneurons. PLoS One 2014; 9:e111105. [PMID: 25347396 PMCID: PMC4210267 DOI: 10.1371/journal.pone.0111105] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 09/28/2014] [Indexed: 11/29/2022] Open
Abstract
A physiological brain function requires neuronal networks to operate within a well-defined range of activity. Indeed, alterations in neuronal excitability have been associated with several pathological conditions, ranging from epilepsy to neuropsychiatric disorders. Changes in inhibitory transmission are known to play a key role in the development of hyperexcitability. However it is largely unknown whether specific interneuronal subpopulations contribute differentially to such pathological condition. In the present study we investigated functional alterations of inhibitory interneurons embedded in a hyperexcitable cortical circuit at the border of chronically induced focal lesions in mouse visual cortex. Interestingly, we found opposite alterations in the excitability of non fast-spiking (Non Fs) and fast-spiking (Fs) interneurons in acute cortical slices from injured animals. Non Fs interneurons displayed a depolarized membrane potential and a higher frequency of spontaneous excitatory postsynaptic currents (sEPSCs). In contrast, Fs interneurons showed a reduced sEPSCs amplitude. The observed downscaling of excitatory synapses targeting Fs interneurons may prevent the recruitment of this specific population of interneurons to the hyperexcitable network. This mechanism is likely to seriously affect neuronal network function and to exacerbate hyperexcitability but it may be important to protect this particular vulnerable population of GABAegic neurons from excitotoxicity.
Collapse
Affiliation(s)
- Barbara Imbrosci
- Institute of Physiology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- * E-mail: (BI); (TM)
| | - Angela Neitz
- Institute of Physiology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Thomas Mittmann
- Institute of Physiology, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- * E-mail: (BI); (TM)
| |
Collapse
|
41
|
Muthuraju S, Islam MR, Pati S, Jaafar H, Abdullah JM, Yusoff KM. Normobaric hyperoxia treatment prevents early alteration in dopamine level in mice striatum after fluid percussion injury: a biochemical approach. Int J Neurosci 2014; 125:686-92. [PMID: 25180987 DOI: 10.3109/00207454.2014.961065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Dopamine (DA) is one of the key neurotransmitters in the striatum, which is functionally important for a variety of cognitive and motor behaviours. It is known that the striatum is vulnerable to damage from traumatic brain injury (TBI). However, a therapeutic approach has not yet been established to treat TBI. Hence, the present work aimed to evaluate the ability of Normobaric hyperoxia treatment (NBOT) to recover dopaminergic neurons following a fluid percussion injury (FPI) as a TBI experimental animal model. To examine this, mice were divided into four groups: (i) Control, (ii) Sham, (iii) FPI and (iv) FPI+NBOT. Mice were anesthetized and surgically prepared for FPI in the striatum and immediate exposure to NBOT at various time points (3, 6, 12 and 24 h). Dopamine levels were then estimated post injury by utilizing a commercially available ELISA method specific to DA. We found that DA levels were significantly reduced at 3 h, but there was no reduction at 6, 12 and 24 h in FPI groups when compared to the control and sham groups. Subjects receiving NBOT showed consistent increased DA levels at each time point when compared with Sham and FPI groups. These results suggest that FPI may alter DA levels at the early post-TBI stages but not in later stages. While DA levels increased in 6, 12 and 24 h in the FPI groups, NBOT could be used to accelerate the prevention of early dopaminergic neuronal damage following FPI injury and improve DA levels consistently.
Collapse
Affiliation(s)
- Sangu Muthuraju
- 1Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| | | | | | | | | | | |
Collapse
|
42
|
Quincozes-Santos A, Bobermin LD, Tramontina AC, Wartchow KM, Tagliari B, Souza DO, Wyse AT, Gonçalves CA. Oxidative stress mediated by NMDA, AMPA/KA channels in acute hippocampal slices: Neuroprotective effect of resveratrol. Toxicol In Vitro 2014; 28:544-51. [DOI: 10.1016/j.tiv.2013.12.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 12/19/2022]
|
43
|
Ferreira APO, Rodrigues FS, Della-Pace ID, Mota BC, Oliveira SM, de Campos Velho Gewehr C, Bobinski F, de Oliveira CV, Brum JS, Oliveira MS, Furian AF, de Barros CSL, dos Santos ARS, Ferreira J, Fighera MR, Royes LFF. HOE-140, an antagonist of B2 receptor, protects against memory deficits and brain damage induced by moderate lateral fluid percussion injury in mice. Psychopharmacology (Berl) 2014; 231:1935-48. [PMID: 24202114 DOI: 10.1007/s00213-013-3336-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/14/2013] [Indexed: 12/14/2022]
Abstract
RATIONALE There are evidences indicating the role of kinins in pathophysiology of traumatic brain injury, but little is known about their action on memory deficits. OBJECTIVES Our aim was to establish the role of bradykinin receptors B₁ (B₁R) and B₂ (B₂R) on the behavioral, biochemical, and histologic features elicited by moderate lateral fluid percussion injury (mLFPI) in mice. METHODS The role of kinin B₁ and B₂ receptors in brain damage, neuromotor, and cognitive deficits induced by mLFPI, was evaluated by means of subcutaneous injection of B₂R antagonist (HOE-140; 1 or 10 nmol/kg) or B₁R antagonist (des-Arg9-[Leu8]-bradykinin (DAL-Bk; 1 or 10 nmol/kg) 30 min and 24 h after brain injury. Brain damage was evaluated in the cortex, being considered as lesion volume, inflammatory, and oxidative damage. The open field and elevated plus maze tests were performed to exclude the nonspecific effects on object recognition memory test. RESULTS Our data revealed that HOE-140 (10 nmol/kg) protected against memory impairment. This treatment attenuated the brain edema, interleukin-1β, tumor necrosis factor-α, and nitric oxide metabolites content elicited by mLFPI. Accordingly, HOE-140 administration protected against the increase of nicotinamide adenine dinucleotide phosphate oxidase activity, thiobarbituric-acid-reactive species, protein carbonylation generation, and Na⁺ K⁺ ATPase inhibition induced by trauma. Histologic analysis showed that HOE-140 reduced lesion volume when analyzed 7 days after brain injury. CONCLUSIONS This study suggests the involvement of the B₂ receptor in memory deficits and brain damage caused by mLFPI in mice.
Collapse
Affiliation(s)
- Ana Paula Oliveira Ferreira
- Laboratório de Bioquímica do Exercício, Departamento de Métodos e Técnicas Desportivas, Centro de Educação Física e Desportos, Universidade Federal de Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brasil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mendes Arent A, de Souza LF, Walz R, Dafre AL. Perspectives on molecular biomarkers of oxidative stress and antioxidant strategies in traumatic brain injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:723060. [PMID: 24689052 PMCID: PMC3943200 DOI: 10.1155/2014/723060] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/04/2013] [Accepted: 12/09/2013] [Indexed: 11/23/2022]
Abstract
Traumatic brain injury (TBI) is frequently associated with abnormal blood-brain barrier function, resulting in the release of factors that can be used as molecular biomarkers of TBI, among them GFAP, UCH-L1, S100B, and NSE. Although many experimental studies have been conducted, clinical consolidation of these biomarkers is still needed to increase the predictive power and reduce the poor outcome of TBI. Interestingly, several of these TBI biomarkers are oxidatively modified to carbonyl groups, indicating that markers of oxidative stress could be of predictive value for the selection of therapeutic strategies. Some drugs such as corticosteroids and progesterone have already been investigated in TBI neuroprotection but failed to demonstrate clinical applicability in advanced phases of the studies. Dietary antioxidants, such as curcumin, resveratrol, and sulforaphane, have been shown to attenuate TBI-induced damage in preclinical studies. These dietary antioxidants can increase antioxidant defenses via transcriptional activation of NRF2 and are also known as carbonyl scavengers, two potential mechanisms for neuroprotection. This paper reviews the relevance of redox biology in TBI, highlighting perspectives for future studies.
Collapse
Affiliation(s)
- André Mendes Arent
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
- Faculty of Medicine, University of South Santa Catarina (Unisul), 88137-270 Palhoça, SC, Brazil
- Neurosurgery Service, São José Regional Hospital (HRSJ-HMG), 88103-901 São José, SC, Brazil
| | - Luiz Felipe de Souza
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
| | - Roger Walz
- Applied Neurosciences Centre (CeNAp) and Department of Medical Clinics, University Hospital, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Alcir Luiz Dafre
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
45
|
Silva LFA, Hoffmann MS, Gerbatin RDR, Fiorin FDS, Dobrachinski F, Mota BC, Wouters ATB, Pavarini SP, Soares FAA, Fighera MR, Royes LFF. Treadmill exercise protects against pentylenetetrazol-induced seizures and oxidative stress after traumatic brain injury. J Neurotrauma 2013; 30:1278-87. [PMID: 23530735 PMCID: PMC3713448 DOI: 10.1089/neu.2012.2577] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of acquired epilepsy, and significant resources are required to develop a better understanding of the pathologic mechanism as targets for potential therapies. Thus, we decided to investigate whether physical exercise after fluid percussion injury (FPI) protects from oxidative and neurochemical alterations as well as from behavioral electroencephalographic (EEG) seizures induced by subeffective convulsive doses of pentylenetetrazol (PTZ; 35 mg/kg). Behavioral and EEG recordings revealed that treadmill physical training increased latency to first clonic and tonic-clonic seizures, attenuated the duration of generalized seizures, and protected against the increase of PTZ-induced Racine scale 5 weeks after neuronal injury. EEG recordings also revealed that physical exercise prevented PTZ-induced amplitude increase in TBI animals. Neurochemical analysis showed that exercise training increased glutathione/oxidized glutathione ratio and glutathione levels per se. Exercise training was also effective against alterations in the redox status, herein characterized by lipid peroxidation (thiobarbituric acid reactive substances), protein carbonyl increase, as well as the inhibition of superoxide dismutase and Na⁺,K⁺-ATPase activities after FPI. On the other hand, histologic analysis with hematoxylin and eosin revealed that FPI induced moderate neuronal damage in cerebral cortex 4 weeks after injury and that physical exercise did not protect against neuronal injury. These data suggest that the ability of physical exercise to reduce FPI-induced seizures is not related to its protection against neuronal damage; however, the effective protection of selected targets, such as Na⁺/K⁺-ATPase elicited by physical exercise, may represent a new line of treatment for post-traumatic seizure susceptibility.
Collapse
Affiliation(s)
- Luiz Fernando Almeida Silva
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brasil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mukherjee S, Zeitouni S, Cavarsan CF, Shapiro LA. Increased seizure susceptibility in mice 30 days after fluid percussion injury. Front Neurol 2013; 4:28. [PMID: 23519723 PMCID: PMC3604640 DOI: 10.3389/fneur.2013.00028] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/03/2013] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) has been reported to increase seizure susceptibility and also contribute to the development of epilepsy. However, the mechanistic basis of the development of increased seizure susceptibility and epilepsy is not clear. Though there is substantial work done using rats, data are lacking regarding the use of mice in the fluid percussion injury (FPI) model. It is unclear if mice, like rats, will experience increased seizure susceptibility following FPI. The availability of a mouse model of increased seizure susceptibility after FPI would provide a basis for the use of genetically modified mice to study mechanism(s) of the development of post-traumatic epilepsy. Therefore, this study was designed to test the hypothesis that, mice subjected to a FPI develop increased seizure susceptibility to a subconvulsive dose of the chemoconvulsant, pentylenetetrazole (PTZ). Three groups of mice were used: FPI, sham, and naïve controls. On day 30 after FPI, mice from the three groups were injected with PTZ. The results showed that FPI mice exhibited an increased severity, frequency, and duration of seizures in response to PTZ injection compared with the sham and naïve control groups. Histopathological assessment was used to characterize the injury at 1, 3, 7, and 30 days after FPI. The results show that mice subjected to the FPI had a pronounced lesion and glial response that was centered at the FPI focus and peaked at 3 days. By 30 days, only minimal evidence of a lesion is observed, although there is evidence of a chronic glial response. These data are the first to demonstrate an early increase in seizure susceptibility following FPI in mice. Therefore, future studies can incorporate transgenic mice into this model to further elucidate mechanisms of TBI-induced increases in seizure susceptibility.
Collapse
Affiliation(s)
- Sanjib Mukherjee
- Department of Surgery, Scott and White Hospital Temple, TX, USA ; Central Texas Veterans Health Care System Temple, TX, USA
| | | | | | | |
Collapse
|
47
|
Saraiva ALL, Ferreira APO, Silva LFA, Hoffmann MS, Dutra FD, Furian AF, Oliveira MS, Fighera MR, Royes LFF. Creatine reduces oxidative stress markers but does not protect against seizure susceptibility after severe traumatic brain injury. Brain Res Bull 2011; 87:180-6. [PMID: 22051612 DOI: 10.1016/j.brainresbull.2011.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 10/04/2011] [Accepted: 10/20/2011] [Indexed: 12/22/2022]
Abstract
Achievements made over the last years have highlighted the important role of creatine in health and disease. However, its effects on hyperexcitable circuit and oxidative damage induced by traumatic brain injury (TBI) are not well understood. In the present study we revealed that severe TBI elicited by fluid percussion brain injury induced oxidative damage characterized by protein carbonylation, thiobarbituric acid reactive species (TBARS) increase and Na(+),K(+)-ATPase activity inhibition 4 and 8 days after neuronal injury. Statistical analysis showed that after TBI creatine supplementation (300 mg/kg, p.o.) decreased the levels of protein carbonyl and TBARS but did not protect against TBI-induced Na(+),K(+)-ATPase activity inhibition. Electroencephalography (EEG) analysis revealed that the injection of a subconvulsant dose of PTZ (35 mg/kg, i.p.), 4 but not 8 days after neuronal injury, decreased latency for the first clonic seizures and increased the time of spent generalized tonic-clonic seizures compared with the sham group. In addition, creatine supplementation had no effect on convulsive parameters induced by a subconvulsant dose of PTZ. Current experiments provide evidence that lipid and protein oxidation represents a separate pathway in the early post-traumatic seizures susceptibility. Furthermore, the lack of consistent anticonvulsant effect exerted by creatine in this early phase suggests that its apparent antioxidant effect does not protect against excitatory input generation induced by TBI.
Collapse
Affiliation(s)
- André Luis Lopes Saraiva
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|