51
|
Tripathi R, Gupta R, Sahu M, Srivastava D, Das A, Ambasta RK, Kumar P. Free radical biology in neurological manifestations: mechanisms to therapeutics interventions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62160-62207. [PMID: 34617231 DOI: 10.1007/s11356-021-16693-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Recent advancements and growing attention about free radicals (ROS) and redox signaling enable the scientific fraternity to consider their involvement in the pathophysiology of inflammatory diseases, metabolic disorders, and neurological defects. Free radicals increase the concentration of reactive oxygen and nitrogen species in the biological system through different endogenous sources and thus increased the overall oxidative stress. An increase in oxidative stress causes cell death through different signaling mechanisms such as mitochondrial impairment, cell-cycle arrest, DNA damage response, inflammation, negative regulation of protein, and lipid peroxidation. Thus, an appropriate balance between free radicals and antioxidants becomes crucial to maintain physiological function. Since the 1brain requires high oxygen for its functioning, it is highly vulnerable to free radical generation and enhanced ROS in the brain adversely affects axonal regeneration and synaptic plasticity, which results in neuronal cell death. In addition, increased ROS in the brain alters various signaling pathways such as apoptosis, autophagy, inflammation and microglial activation, DNA damage response, and cell-cycle arrest, leading to memory and learning defects. Mounting evidence suggests the potential involvement of micro-RNAs, circular-RNAs, natural and dietary compounds, synthetic inhibitors, and heat-shock proteins as therapeutic agents to combat neurological diseases. Herein, we explain the mechanism of free radical generation and its role in mitochondrial, protein, and lipid peroxidation biology. Further, we discuss the negative role of free radicals in synaptic plasticity and axonal regeneration through the modulation of various signaling molecules and also in the involvement of free radicals in various neurological diseases and their potential therapeutic approaches. The primary cause of free radical generation is drug overdosing, industrial air pollution, toxic heavy metals, ionizing radiation, smoking, alcohol, pesticides, and ultraviolet radiation. Excessive generation of free radicals inside the cell R1Q1 increases reactive oxygen and nitrogen species, which causes oxidative damage. An increase in oxidative damage alters different cellular pathways and processes such as mitochondrial impairment, DNA damage response, cell cycle arrest, and inflammatory response, leading to pathogenesis and progression of neurodegenerative disease other neurological defects.
Collapse
Affiliation(s)
- Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India.
- , Delhi, India.
- Molecular Neuroscience and Functional Genomics Laboratory, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
52
|
Perspectives on the Molecular Mediators of Oxidative Stress and Antioxidant Strategies in the Context of Neuroprotection and Neurolongevity: An Extensive Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7743705. [PMID: 36062188 PMCID: PMC9439934 DOI: 10.1155/2022/7743705] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/09/2022] [Indexed: 12/11/2022]
Abstract
Molecules with at least one unpaired electron in their outermost shell are known as free radicals. Free radical molecules are produced either within our bodies or by external sources such as ozone, cigarette smoking, X-rays, industrial chemicals, and air pollution. Disruption of normal cellular homeostasis by redox signaling may result in cardiovascular, neurodegenerative diseases and cancer. Although ROS (reactive oxygen species) are formed in the GI tract, little is known about how they contribute to pathophysiology and disease etiology. When reactive oxygen species and antioxidants are in imbalance in our bodies, they can cause cell structure damage, neurodegenerative diseases, diabetes, hypercholesterolemia, atherosclerosis, cancer, cardiovascular diseases, metabolic disorders, and other obesity-related disorders, as well as protein misfolding, mitochondrial dysfunction, glial cell activation, and subsequent cellular apoptosis. Neuron cells are gradually destroyed in neurodegenerative diseases. The production of inappropriately aggregated proteins is strongly linked to oxidative stress. This review's goal is to provide as much information as possible about the numerous neurodegenerative illnesses linked to oxidative stress. The possibilities of multimodal and neuroprotective therapy in human illness, using already accessible medications and demonstrating neuroprotective promise in animal models, are highlighted. Neuroprotection and neurolongevity may improve from the use of bioactive substances from medicinal herbs like Allium stadium, Celastrus paniculatus, and Centella asiatica. Many neuroprotective drugs' possible role has been addressed. Preventing neuroinflammation has been demonstrated in several animal models.
Collapse
|
53
|
Methamphetamine induced neurotoxic diseases, molecular mechanism, and current treatment strategies. Biomed Pharmacother 2022; 154:113591. [PMID: 36007276 DOI: 10.1016/j.biopha.2022.113591] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Methamphetamine (MA) is a extremely addictive psychostimulant drug with a significant abuse potential. Long-term MA exposure can induce neurotoxic effects through oxidative stress, mitochondrial functional impairment, endoplasmic reticulum stress, the activation of astrocytes and microglial cells, axonal transport barriers, autophagy, and apoptosis. However, the molecular and cellular mechanisms underlying MA-induced neurotoxicity remain unclear. MA abuse increases the chances of developing neurotoxic conditions such as Parkinson's disease (PD), Alzheimer's disease (AD) and other neurotoxic diseases. MA increases the risk of PD by increasing the expression of alpha-synuclein (ASYN). Furthermore, MA abuse is linked to high chances of developing AD and subsequent neurodegeneration due to biological variations in the brain region or genetic and epigenetic variations. To date, there is no Food and Drug Administration (FDA)-approved therapy for MA-induced neurotoxicity, although many studies are being conducted to develop effective therapeutic strategies. Most current studies are now focused on developing therapies to diminish the neurotoxic effects of MA, based on the underlying mechanism of neurotoxicity. This review article highlights current research on several therapeutic techniques targeting multiple pathways to reduce the neurotoxic effects of MA in the brain, as well as the putative mechanism of MA-induced neurotoxicity.
Collapse
|
54
|
Aboutabl ME, Elkhateeb WA, Masoud MA, Daba GM, Afifi AH, Hussein RA. HPLC and GC-MS based metabolic profiles and in vivo anticonvulsant, sedative, and antinociceptive potentials of truffles Tirmania nivea and Tirmania pinoyi hydromethanolic extracts in mice. Biomed Chromatogr 2022; 36:e5481. [PMID: 35971328 DOI: 10.1002/bmc.5481] [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: 06/05/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022]
Abstract
GC-MS and HPLC analyses of the hydromethanolic extracts of the truffles Tirmania nivea (TN) and Tirmania pinoyi (TP) revealed the presence of 18 metabolites and 11 polyphenols, respectively. In vivo, TP extract protected against subcutaneous pentylenetetrazole (scPTZ) and maximal electric shock (MES)-induced convulsions faster than TN. TP (100 and 300 mg/kg) showed 100% protection and longer duration than TN in the scPTZ test. Similarly, at 300 mg/kg, TP demonstrated a quicker start (75%) and longer duration of action (100%) than TN in MES test. In scPTZ test, ED50 of TP demonstrated greater anticonvulsant efficacy than TN. In mice given TP and TN treatments, the brain GABA levels were noticeably increased. TP (100 and 300mg/kg) produced a notable sedative effect in open field test, whereas TN (100 or 300 mg/kg) and TP (300 mg/kg) reduced sleep latency by 79, 52, and 45%, respectively. In writhing test, TN (100 or 300mg/kg) significantly enhanced analgesic efficacy by 50 and 87%, respectively. Comparatively, in formalin test, TP and TN at a dosage of 300 mg/kg decreased the length of the licking by 34 and 59%, respectively. For the first time, this study explains the anticonvulsant, sedative, central, and peripheral analgesic activities of truffle extracts.
Collapse
Affiliation(s)
- Mona E Aboutabl
- Medicinal and Pharmaceutical Chemistry Department (Pharmacology Group), Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Waill A Elkhateeb
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Marwa A Masoud
- Pharmacology Department, National Organization for Drug Control and Research, Egyptian Drug Authority (EDA), Giza, Egypt
| | - Ghoson M Daba
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Ahmed H Afifi
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| | - Rehab A Hussein
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), Giza, Egypt
| |
Collapse
|
55
|
Oliveira MET, Paulino GVB, Dos Santos Júnior ED, da Silva Oliveira FA, Melo VMM, Ursulino JS, de Aquino TM, Shetty AK, Landell MF, Gitaí DLG. Multi-omic Analysis of the Gut Microbiome in Rats with Lithium-Pilocarpine-Induced Temporal Lobe Epilepsy. Mol Neurobiol 2022; 59:6429-6446. [PMID: 35962889 DOI: 10.1007/s12035-022-02984-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 07/29/2022] [Indexed: 11/25/2022]
Abstract
Evidence supports that the gut microbiota and bacteria-dependent metabolites influence the maintenance of epileptic brain activity. However, the alterations in the gut microbiota between epileptic versus healthy individuals are poorly understood. We used a multi-omic approach to evaluate the changes in the composition of gut metagenome as well in the fecal metabolomic profile in rats before and after being submitted to status epilepticus (SE)-induced temporal lobe epilepsy (TLE). The 16S ribosomal RNA (rRNA) sequencing of fecal samples coupled to bioinformatic analysis revealed taxonomic, compositional, and functional shifts in epileptic rats. The species richness (Chao1 index) was significantly lower in the post-TLE group, and the β-diversity analysis revealed clustering separated from the pre-TLE group. The taxonomic abundance analysis showed a significant increase of phylum Desulfobacterota and a decrease of Patescibacteria in the post-TLE group. The DESEq2 and LEfSe analysis resulted in 18 genera significantly enriched between post-TLE and pre-TLE groups at the genus level. We observed that epileptic rats present a peculiar metabolic phenotype, including a lower concentration of D-glucose and L-lactic acid and a higher concentration of L-glutamic acid and glycine. The microbiota-host metabolic correlation analysis showed that the genera differentially abundant in post-TLE rats are associated with the altered metabolites, especially the proinflammatory Desulfovibrio and Marvinbryantia, which were enriched in epileptic animals and positively correlated with these excitatory neurotransmitters and carbohydrate metabolites. Therefore, our data revealed a correlation between dysbacteriosis in epileptic animals and fecal metabolites that are known to be relevant for maintaining epileptic brain activity by enhancing chronic inflammation, an excitatory-inhibitory imbalance, and/or a metabolic disturbance. These data are promising and suggest that targeting the gut microbiota could provide a novel avenue for preventing and treating acquired epilepsy. However, the causal relationship between these microbial/metabolite components and the SRS occurrence still needs further exploration.
Collapse
Affiliation(s)
- Maria Eduarda T Oliveira
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Gustavo V B Paulino
- Laboratory of Molecular Diversity (LDM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Erivaldo D Dos Santos Júnior
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Francisca A da Silva Oliveira
- Laboratory of Microbial Ecology and Biotechnology (Lembiotech), Department of Biology, Universidade Federal Do Ceará, Campus do Pici, Bloco 909, Fortaleza, CE, 60455-760, Brazil
| | - Vânia M M Melo
- Laboratory of Microbial Ecology and Biotechnology (Lembiotech), Department of Biology, Universidade Federal Do Ceará, Campus do Pici, Bloco 909, Fortaleza, CE, 60455-760, Brazil
| | - Jeferson S Ursulino
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Thiago M de Aquino
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, TX, USA
| | - Melissa Fontes Landell
- Laboratory of Molecular Diversity (LDM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil.
| | - Daniel Leite Góes Gitaí
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil.
| |
Collapse
|
56
|
Research progress on oxidative stress regulating different types of neuronal death caused by epileptic seizures. Neurol Sci 2022; 43:6279-6298. [DOI: 10.1007/s10072-022-06302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/24/2022] [Indexed: 12/09/2022]
|
57
|
Manavi MA. Neuroprotective effects of glucagon-like peptide-1 (GLP-1) analogues in epilepsy and associated comorbidities. Neuropeptides 2022; 94:102250. [PMID: 35561568 DOI: 10.1016/j.npep.2022.102250] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Abstract
Epilepsy is a common neurological condition induced by losing equilibrium of different pathway as well as neurotransmitters that affects over 50 million people globally. Furthermore, long-term administration of anti-seizure medications has been associated with psychological adverse effects. Also, epilepsy has been related to an increased prevalence of obesity and called type 2 diabetes mellitus. On the other hand, GLP-1 receptors are located throughout the brain, including the hippocampus, which have been associated to majority of neurological conditions, such as epilepsy and psychiatric disorders. Moreover, the impact of different GLP-1 analogues on diverse neurotransmitter systems and associated cellular and molecular pathways as a potential therapeutic target for epilepsy and associated comorbidities has piqued curiosity. In this regard, the anticonvulsant effects of GLP-1 analogues have been investigated in various animal models and promising results such as anticonvulsants as well as cognitive improvements have been observed. For instance, GLP-1 analogues like liraglutide in addition to their possible anticonvulsant benefits, could be utilized to alleviate mental cognitive problems caused by both epilepsy and anti-seizure medication side effects. In this review and growing protective function of GLP-1 in epilepsy induced by disturbed neurotransmitter pathways and the probable mechanisms of action of GLP-1 analogues as well as the GLP-1 receptor in these effects have been discussed.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
58
|
Vicente-Silva W, Silva-Freitas FR, Beserra-Filho JIA, Cardoso GN, Silva-Martins S, Sarno TA, Silva SP, Soares-Silva B, Dos Santos JR, da Silva RH, Prado CM, Ueno AK, Lago JHG, Ribeiro AM. Sakuranetin exerts anticonvulsant effect in bicuculline-induced seizures. Fundam Clin Pharmacol 2022; 36:663-673. [PMID: 35156229 DOI: 10.1111/fcp.12768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/07/2023]
Abstract
Epilepsy is a chronic neurological disorder characterized by an abnormal, spontaneous, and synchronized neuronal hyperactivity. Therapeutic approaches for controlling epileptic seizures are associated with pharmacoresistance and side effects burden. Previous studies reported that different natural products may have neuroprotector effects. Sakuranetin (SAK) is a flavanone with antiparasitic, anti-inflammatory, antimutagenic, antiallergic, and antioxidant activity. In the present work, the effect of SAK on seizures in a model of status epilepticus induced by bicuculline (BIC) in mice was evaluated. Male Swiss mice received an intracerebroventricular injection (i.c.v.) of SAK (1, 10, or 20 mg/kg-SAK1, SAK10, or SAK20). Firstly, animals were evaluated in the open field (OF; 20 min), afterwards in the elevated plus maze (EPM) test (5 min). Next, 30 min prior the administration of BIC (1 mg/kg), mice received an injection of SAK (1 or 10 mg/kg, i.c.v.) and were observed in the OF (20 min) for seizures assessment. After behavioral procedures, immunohistochemical analysis of c-Fos was performed. Our main results showed that the lowest doses of SAK (1 and 10 mg/kg) increased the total distance traveled in the OF, moreover protected against seizures and death on the BIC-induced seizures model. Furthermore, SAK treatment reduced neuronal activity on the dentate gyrus of the BIC-treated animals. Taken together, our results suggest an anticonvulsant effect of SAK, which could be used for the development of anticonvulsants based on natural products from herbal source.
Collapse
Affiliation(s)
- Wilson Vicente-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | | | | | - Suellen Silva-Martins
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Tamires Alves Sarno
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Sara Pereira Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Beatriz Soares-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | - Regina Helena da Silva
- Department of Pharmacology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Carla Máximo Prado
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Anderson Keity Ueno
- Department of Biosciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | | | | |
Collapse
|
59
|
Ranasinghe R, Mathai M, Zulli A. Revisiting the therapeutic potential of tocotrienol. Biofactors 2022; 48:813-856. [PMID: 35719120 PMCID: PMC9544065 DOI: 10.1002/biof.1873] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The therapeutic potential of the tocotrienol group stems from its nutraceutical properties as a dietary supplement. It is largely considered to be safe when consumed at low doses for attenuating pathophysiology as shown by animal models, in vitro assays, and ongoing human trials. Medical researchers and the allied sciences have experimented with tocotrienols for many decades, but its therapeutic potential was limited to adjuvant or concurrent treatment regimens. Recent studies have focused on targeted drug delivery by enhancing the bioavailability through carriers, self-sustained emulsions, nanoparticles, and ethosomes. Epigenetic modulation and computer remodeling are other means that will help increase chemosensitivity. This review will focus on the systemic intracellular anti-cancer, antioxidant, and anti-inflammatory mechanisms that are stimulated and/or regulated by tocotrienols while highlighting its potent therapeutic properties in a diverse group of clinical diseases.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Michael Mathai
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Anthony Zulli
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| |
Collapse
|
60
|
Alfawaz HA, El-Ansary A, Al-Ayadhi L, Bhat RS, Hassan WM. Protective Effects of Bee Pollen on Multiple Propionic Acid-Induced Biochemical Autistic Features in a Rat Model. Metabolites 2022; 12:metabo12070571. [PMID: 35888695 PMCID: PMC9323335 DOI: 10.3390/metabo12070571] [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: 06/06/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 11/26/2022] Open
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental disorders that clinically presented as impaired social interaction, repetitive behaviors, and weakened communication. The use of bee pollen as a supplement rich in amino acids amino acids, vitamins, lipids, and countless bioactive substances may lead to the relief of oxidative stress, neuroinflammation, glutamate excitotoxicity, and impaired neurochemistry as etiological mechanisms autism. Thirty young male Western albino rats were randomly divided as: Group I-control; Group II, in which autism was induced by the oral administration of 250 mg propionic acid/kg body weight/day for three days followed by orally administered saline until the end of experiment and Group III, the bee pollen-treated group, in which the rats were treated with 250 mg/kg body weight of bee pollen for four weeks before autism was induced as described for Group II. Markers related to oxidative stress, apoptosis, inflammation, glutamate excitotoxicity, and neurochemistry were measured in the brain tissue. Our results indicated that while glutathione serotonin, dopamine, gamma-aminobutyric acid (GABA), GABA/Glutamate ratio, and vitamin C were significantly reduced in propionic acid-treated group (p < 0.05), glutamate, IFN-γ, IL-1A, IL-6, caspase-3, and lipid peroxide levels were significantly elevated (p < 0.05). Bee pollen supplementation demonstrates protective potency presented as amelioration of most of the measured variables with significance range between (p < 0.05)−(p < 0.001).
Collapse
Affiliation(s)
- Hanan A. Alfawaz
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Afaf El-Ansary
- Central Research Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, Riyadh 11495, Saudi Arabia
- Correspondence: ; Tel.: +966-508462529; Fax: +966-11-4682184
| | - Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Sciences, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Wail M. Hassan
- Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA;
| |
Collapse
|
61
|
Contreras-García IJ, Cárdenas-Rodríguez N, Romo-Mancillas A, Bandala C, Zamudio SR, Gómez-Manzo S, Hernández-Ochoa B, Mendoza-Torreblanca JG, Pichardo-Macías LA. Levetiracetam Mechanisms of Action: From Molecules to Systems. Pharmaceuticals (Basel) 2022; 15:ph15040475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic disease that affects millions of people worldwide. Antiepileptic drugs (AEDs) are used to control seizures. Even though parts of their mechanisms of action are known, there are still components that need to be studied. Therefore, the search for novel drugs, new molecular targets, and a better understanding of the mechanisms of action of existing drugs is still crucial. Levetiracetam (LEV) is an AED that has been shown to be effective in seizure control and is well-tolerable, with a novel mechanism of action through an interaction with the synaptic vesicle protein 2A (SV2A). Moreover, LEV has other molecular targets that involve calcium homeostasis, the GABAergic system, and AMPA receptors among others, that might be integrated into a single mechanism of action that could explain the antiepileptogenic, anti-inflammatory, neuroprotective, and antioxidant properties of LEV. This puts it as a possible multitarget drug with clinical applications other than for epilepsy. According to the above, the objective of this work was to carry out a comprehensive and integrative review of LEV in relation to its clinical uses, structural properties, therapeutical targets, and different molecular, genetic, and systemic action mechanisms in order to consider LEV as a candidate for drug repurposing.
Collapse
Affiliation(s)
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico;
| | - Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| | - Julieta Griselda Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| |
Collapse
|
62
|
Distinct Epileptogenic Mechanisms Associated with Seizures in Wolf-Hirschhorn Syndrome. Mol Neurobiol 2022; 59:3159-3169. [DOI: 10.1007/s12035-022-02792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022]
|
63
|
Batista MA, de Lima Teixeira dos Santos AVT, do Nascimento AL, Moreira LF, Souza IRS, da Silva HR, Pereira ACM, da Silva Hage-Melim LI, Carvalho JCT. Potential of the Compounds from Bixa orellana Purified Annatto Oil and Its Granules (Chronic ®) against Dyslipidemia and Inflammatory Diseases: In Silico Studies with Geranylgeraniol and Tocotrienols. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27051584. [PMID: 35268686 PMCID: PMC8911567 DOI: 10.3390/molecules27051584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022]
Abstract
Some significant compounds present in annatto are geranylgeraniol and tocotrienols. These compounds have beneficial effects against hyperlipidemia and chronic diseases, where oxidative stress and inflammation are present, but the exact mechanism of action of such activities is still a subject of research. This study aimed to evaluate possible mechanisms of action that could be underlying the activities of these molecules. For this, in silico approaches such as ligand topology (PASS and SEA servers) and molecular docking with the software GOLD were used. Additionally, we screened some pharmacokinetic and toxicological parameters using the servers PreADMET, SwissADME, and ProTox-II. The results corroborate the antidyslipidemia and anti-inflammatory activities of geranylgeraniol and tocotrienols. Notably, some new mechanisms of action were predicted to be potentially underlying the activities of these compounds, including inhibition of squalene monooxygenase, lanosterol synthase, and phospholipase A2. These results give new insight into new mechanisms of action involved in these molecules from annatto and Chronic®.
Collapse
Affiliation(s)
- Mateus Alves Batista
- Laboratory of Pharmaceutical and Medicinal Chemistry (PharMedChem), Federal University of Amapá, Amapá, Macapá 68902-280, Brazil; (M.A.B.); (L.I.d.S.H.-M.)
| | - Abrahão Victor Tavares de Lima Teixeira dos Santos
- Laboratory of Drugs Research, Biology and Healthy Sciences Department, Pharmacy Faculty, Federal University of Amapá, Rod. JK, km 02, Amapá, Macapá 68902-280, Brazil; (A.V.T.d.L.T.d.S.); (A.L.d.N.); (L.F.M.); (H.R.d.S.)
| | - Aline Lopes do Nascimento
- Laboratory of Drugs Research, Biology and Healthy Sciences Department, Pharmacy Faculty, Federal University of Amapá, Rod. JK, km 02, Amapá, Macapá 68902-280, Brazil; (A.V.T.d.L.T.d.S.); (A.L.d.N.); (L.F.M.); (H.R.d.S.)
| | - Luiz Fernando Moreira
- Laboratory of Drugs Research, Biology and Healthy Sciences Department, Pharmacy Faculty, Federal University of Amapá, Rod. JK, km 02, Amapá, Macapá 68902-280, Brazil; (A.V.T.d.L.T.d.S.); (A.L.d.N.); (L.F.M.); (H.R.d.S.)
| | - Indira Ramos Senna Souza
- Diamantina Chapada Regional Hospital, Avenida Francisco Costa, 350-468, Vasco Filho, Bahia, Seabra 46900-000, Brazil;
| | - Heitor Ribeiro da Silva
- Laboratory of Drugs Research, Biology and Healthy Sciences Department, Pharmacy Faculty, Federal University of Amapá, Rod. JK, km 02, Amapá, Macapá 68902-280, Brazil; (A.V.T.d.L.T.d.S.); (A.L.d.N.); (L.F.M.); (H.R.d.S.)
| | - Arlindo César Matias Pereira
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), São Paulo, Ribeirão Preto 05508-000, Brazil;
| | - Lorane Izabel da Silva Hage-Melim
- Laboratory of Pharmaceutical and Medicinal Chemistry (PharMedChem), Federal University of Amapá, Amapá, Macapá 68902-280, Brazil; (M.A.B.); (L.I.d.S.H.-M.)
| | - José Carlos Tavares Carvalho
- Laboratory of Drugs Research, Biology and Healthy Sciences Department, Pharmacy Faculty, Federal University of Amapá, Rod. JK, km 02, Amapá, Macapá 68902-280, Brazil; (A.V.T.d.L.T.d.S.); (A.L.d.N.); (L.F.M.); (H.R.d.S.)
- Correspondence:
| |
Collapse
|
64
|
Shou JW, Shaw PC. Therapeutic Efficacies of Berberine against Neurological Disorders: An Update of Pharmacological Effects and Mechanisms. Cells 2022; 11:cells11050796. [PMID: 35269418 PMCID: PMC8909195 DOI: 10.3390/cells11050796] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Neurological disorders are ranked as the leading cause of disability and the second leading cause of death worldwide, underscoring an urgent necessity to develop novel pharmacotherapies. Berberine (BBR) is a well-known phytochemical isolated from a number of medicinal herbs. BBR has attracted much interest for its broad range of pharmacological actions in treating and/or managing neurological disorders. The discoveries in basic and clinical studies of the effects of BBR on neurological disorders in the last decade have provided novel evidence to support the potential therapeutical efficacies of BBR in treating neurological diseases. In this review, we summarized the pharmacological properties and therapeutic applications of BBR against neurological disorders in the last decade. We also emphasized the major pathways modulated by BBR, which provides firm evidence for BBR as a promising drug candidate for neurological disorders.
Collapse
Affiliation(s)
- Jia-Wen Shou
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 852852, China;
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 852852, China;
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 852852, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants and Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 852852, China
- Correspondence:
| |
Collapse
|
65
|
Zaaboul F, Liu Y. Vitamin E in foodstuff: Nutritional, analytical, and food technology aspects. Compr Rev Food Sci Food Saf 2022; 21:964-998. [PMID: 35181987 DOI: 10.1111/1541-4337.12924] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 11/21/2021] [Accepted: 01/10/2022] [Indexed: 12/16/2022]
Abstract
Vitamin E is a group of isoprenoid chromanols with different biological activities. It comprises eight oil-soluble compounds: four tocopherols, namely, α-, β-, γ-, and δ-tocopherols; and four tocotrienols, namely, α-, β-, γ, and δ-tocotrienols. Vitamin E isomers are well-known for their antioxidant activity, gene-regulation effects, and anti-inflammatory and nephroprotective properties. Considering that vitamin E is exclusively synthesized by photosynthetic organisms, animals can only acquire it through their diet. Plant-based food is the primary source of vitamin E; hence, oils, nuts, fruits, and vegetables with high contents of vitamin E are mostly consumed after processing, including industrial processes and home-cooking, which involve vitamin E profile and content alteration during their preparation. Accordingly, it is essential to identify the vitamin E content and profile in foodstuff to match daily intake requirements. This review summarizes recent advances in vitamin E chemistry, metabolism and metabolites, current knowledge on their contents and profiles in raw and processed plant foods, and finally, their modern developments in analytical methods.
Collapse
Affiliation(s)
- Farah Zaaboul
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic China
| | - YuanFa Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic China
| |
Collapse
|
66
|
Pardo-Peña K, Yañez-Hernández A, Medina-Ceja L, Morales-Villagrán A. Ellagic acid and allopurinol decrease H 2O 2 concentrations, epileptiform activity and astrogliosis after status epilepticus in the hippocampus of adult rats. Exp Brain Res 2022; 240:1191-1203. [PMID: 35171306 DOI: 10.1007/s00221-022-06323-9] [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: 03/03/2021] [Accepted: 02/04/2022] [Indexed: 11/04/2022]
Abstract
Status epilepticus (SE) can result in an overproduction of hydrogen peroxide (H2O2), which contributes to oxidative stress and brain injury during different phases of epileptogenesis and seizures. The purpose of this study was to evaluate the effects of ellagic acid and allopurinol administered after SE on H2O2 concentrations, electrical activity and GFAP immunoreactivity in the hippocampus of rats evaluated on Day 18 after SE. H2O2 levels were measured using an online technique with high temporal resolution and simultaneous electrical activity recording. For this purpose, the lateral ventricles of male Wistar rats (200-250 g) were injected with pilocarpine (2.4 mg/2 µl) to induce SE. After SE, rats were injected with ellagic acid (50 mg/kg i.p., and two additional doses at 24 and 48 h) or allopurinol (50 mg/kg i.p., single dose). Administration of ellagic acid or allopurinol after SE significantly reduced the H2O2 concentrations and decreased the presence of epileptiform activity and GFAP immunoreactivity in the hippocampus 18 days after SE. In conclusion, the administration of antioxidants potentially reduces oxidative stress, which indicates the possible attenuation of the neurobiological consequences after SE.
Collapse
Affiliation(s)
- Kenia Pardo-Peña
- Laboratory of Neurophysiology, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Camino Ing. Ramón Padilla Sánchez 2100, Las Agujas, Nextipac, Zapopan, 45200, Jalisco, Mexico.
| | - Aldo Yañez-Hernández
- Laboratory of Neurophysiology, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Camino Ing. Ramón Padilla Sánchez 2100, Las Agujas, Nextipac, Zapopan, 45200, Jalisco, Mexico
| | - Laura Medina-Ceja
- Laboratory of Neurophysiology, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Camino Ing. Ramón Padilla Sánchez 2100, Las Agujas, Nextipac, Zapopan, 45200, Jalisco, Mexico
| | | |
Collapse
|
67
|
Ishola IO, Akinleye MO, Afolayan OO, Okonkwo HE, Animashaun OT, Agbaje EO. Anticonvulsant Activity of Nymphaea Lotus Linn. Extract in Mice: the Role of GABAergic-glutamatergic Neurotransmission and Antioxidant Defense Mechanisms. Epilepsy Res 2022; 181:106871. [DOI: 10.1016/j.eplepsyres.2022.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 11/16/2022]
|
68
|
Mishra NK, Engel J, Liebeskind DS, Sharma VK, Hirsch LJ, Kasner SE, French JA, Devinsky O, Friedman A, Dawson J, Quinn TJ, Selim M, de Havenon A, Yasuda CL, Cendes F, Benninger F, Zaveri HP, Burneo JG, Srivastava P, Bhushan Singh M, Bhatia R, Vishnu VY, Bentes C, Ferro J, Weiss S, Sivaraju A, Kim JA, Galovic M, Gilmore EJ, Pitkänen A, Davis K, Sansing LH, Sheth KN, Paz JT, Singh A, Sheth S, Worrall BB, Grotta JC, Casillas-Espinos PM, Chen Z, Nicolo JP, Yan B, Kwan P. International Post Stroke Epilepsy Research Consortium (IPSERC): A consortium to accelerate discoveries in preventing epileptogenesis after stroke. Epilepsy Behav 2022; 127:108502. [PMID: 34968775 DOI: 10.1016/j.yebeh.2021.108502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/18/2022]
Affiliation(s)
| | - Jerome Engel
- Department of Neurology, University of California Los Angeles, Los Angeles, USA
| | - David S Liebeskind
- Department of Neurology, University of California Los Angeles, Los Angeles, USA
| | - Vijay K Sharma
- YLL School of Medicine, National University of Singapore and Division of Neurology, National University Health System, Singapore
| | | | - Scott E Kasner
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Jacqueline A French
- Department of Neurology, NYU Grossman School of Medicine, New York City, USA
| | - Orrin Devinsky
- Department of Neurology, NYU Grossman School of Medicine, New York City, USA
| | - Alon Friedman
- Department of Brain and Cognitive Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Jesse Dawson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK
| | - Terence J Quinn
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK
| | - Magdy Selim
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | | | - Clarissa L Yasuda
- Department of Neurology, School of Medical Sciences, University of Campinas - UNICAMP, Sao Paulo, Brazil
| | - Fernando Cendes
- Department of Neurology, School of Medical Sciences, University of Campinas - UNICAMP, Sao Paulo, Brazil
| | - Felix Benninger
- Department of Neurology, Rabin Medical Center, Tel Aviv, Israel
| | | | - Jorge G Burneo
- Epilepsy Program, Department of Clinical Neurological Sciences, and Neuroepidemiology Unit, Western University, London, Ontario, Canada
| | - Padma Srivastava
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Mamta Bhushan Singh
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Bhatia
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - V Y Vishnu
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Carla Bentes
- Department of Neurology, University of Lisboa, Lisbon, Portugal
| | - Jose Ferro
- Department of Neurology, University of Lisboa, Lisbon, Portugal
| | - Shennan Weiss
- Department of Neurology, State University of New York (SUNY) Downstate, NY, USA
| | | | - Jennifer A Kim
- Department of Neurology, Yale University, New Haven, USA
| | - Marian Galovic
- Department of Neurology, University of Zurich, Zurich, Switzerland
| | | | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kathryn Davis
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | | - Kevin N Sheth
- Department of Neurology, Yale University, New Haven, USA
| | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, USA; Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Anuradha Singh
- Department of Neurology, Icahn School of Medicine at Mt. Sinai, NY, USA
| | - Sunil Sheth
- Department of Neurology, University of Texas Health Sciences Center, Houston, USA
| | - Bradford B Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia, Charlottesville, USA
| | - James C Grotta
- Department of Neurology, Memorial-Hermann Texas Medical Center, Houston, USA
| | - Pablo M Casillas-Espinos
- Department of Neuroscience, Monash University, Alfred Hospital, Melbourne, Australia; Departments of Neurology and Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Zhibin Chen
- Department of Neuroscience, Monash University, Alfred Hospital, Melbourne, Australia; Departments of Neurology and Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - John-Paul Nicolo
- Department of Neuroscience, Monash University, Alfred Hospital, Melbourne, Australia; Departments of Neurology and Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Bernard Yan
- Departments of Neurology and Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Patrick Kwan
- Department of Neuroscience, Monash University, Alfred Hospital, Melbourne, Australia; Departments of Neurology and Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia.
| |
Collapse
|
69
|
Alvi AM, Shah FA, Muhammad AJ, Feng J, Li S. 1,3,4, Oxadiazole Compound A3 Provides Robust Protection Against PTZ-Induced Neuroinflammation and Oxidative Stress by Regulating Nrf2-Pathway. J Inflamm Res 2022; 14:7393-7409. [PMID: 35002275 PMCID: PMC8721032 DOI: 10.2147/jir.s333451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Background Epilepsy is a common neurological disorder that is characterized by recurrent episodes of seizures. Various studies have demonstrated a direct association between oxidative stress and inflammation in several neurological disorders including epilepsy. This study aimed to investigate the neuroprotective effects of a synthetic 1,3,4, oxadiazole compound A3 against pentylenetetrazole (PTZ)-induced kindling and seizure model. Methodology PTZ was administered in a sub-convulsive dose of 40 mg/kg for 15 days, at 48-hour intervals to male Swiss-Albino mice until animals were fully kindled. Two different doses of A3 (10 mg/kg and 30 mg/kg) were administered to find out the effective dose of A3 and to further demonstrate the relative role of nuclear factor E2-related factor (Nrf2) in the PTZ-induced kindled model. Results Our results demonstrated a compromised antioxidant capacity associated with a low level of catalase (CAT), superoxide dismutase (SOD), glutathione (GST), and glutathione S-transferase (GSH) in the kindled group. However, the PTZ-induced group demonstrated an elevated level of lipid peroxidation (LPO) level parallel to pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), mediators as cyclooxygenase (COX-2), and nuclear factor kappa B (NFκB). Furthermore, the A3 treatment reversed these changes and overexpressed the antioxidant Nrf2 gene and its downstream HO-1. To further investigate the involvement of Nrf2, we employed an Nrf2-inhibitor, ie, all-trans retinoic acid (ATRA), that further aggravated the PTZ toxicity. Moreover, vascular endothelial growth factor (VEGF) expression was evaluated to assess the extent of BBB disruption. Conclusion The findings of this study suggest that A3 could mediate neuroprotection possibly by activating Nrf2 dependent downregulation of inflammatory cascades.
Collapse
Affiliation(s)
- Arooj Mohsin Alvi
- Department of Neonatology, Shenzhen Children's Hospital Shenzhen, Shenzhen, People's Republic of China.,Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Asmaa Jan Muhammad
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children's Hospital Shenzhen, Shenzhen, People's Republic of China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People's Republic of China
| |
Collapse
|
70
|
Jiang H, Zhang S. Therapeutic effect of acute and chronic use of different doses of vitamin D3 on seizure responses and cognitive impairments induced by pentylenetetrazole in immature male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:84-95. [PMID: 35656438 PMCID: PMC9118278 DOI: 10.22038/ijbms.2021.60123.13328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022]
Abstract
Objectives This study aimed to evaluate the effects of acute and chronic intake of different doses of vitamin D3 on seizure responses and cognitive impairment induced by pentylenetetrazole (PTZ) in immature male rats. Materials and Methods Sixty-six immature male NMRI rats were divided into control (10), epileptic (10), and treatment groups (46). The stage 5 latency (S5L) and stage 5 duration (S5D) were assessed along with the shuttle box test. Levels of antioxidant enzymes and inflammatory factors along with genes involved in inflammation, oxidative damage, apoptosis, and mTORc1 were measured in the hippocampus tissue of the brain of controlled and treated rats. Serum levels of parathyroid hormone (PTH), vitamin D, calcium, and phosphorus were also assessed. Results The results showed that the ability to learn, memory consolidation, and memory retention in epileptic rats were reduced. In addition, S5D increased and S5L decreased in epileptic rats, while being effectively ameliorated by chronic and acute vitamin D intake. The results showed that vitamin D in different doses acutely and chronically decreased the levels of oxidative and inflammatory biomarkers in hippocampus tissue and inhibited the expression of genes involved in inflammation, oxidative damage, apoptosis, and mTORc1 in the hippocampus tissue of epileptic rats. Conclusion The results showed that vitamin D in different doses acutely and chronically could improve cognitive impairments and convulsive responses in epileptic rats by improving neurotransmission, inflammation, apoptosis, and oxidative damage.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Pediatric, Weinan Maternal and Child Health Hospital, Weinan, 714000, China
| | - Suying Zhang
- Department of Child Health, Weinan Central Hospital, Weinan, 714000, China
| |
Collapse
|
71
|
Arida RM, Passos AA, Graciani AL, Brogin JAF, Ribeiro MDAL, Faber J, Gutierre RC, Teixeira-Machado L. The Potential Role of Previous Physical Exercise Program to Reduce Seizure Susceptibility: A Systematic Review and Meta-Analysis of Animal Studies. Front Neurol 2021; 12:771123. [PMID: 34956052 PMCID: PMC8702853 DOI: 10.3389/fneur.2021.771123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/26/2021] [Indexed: 12/09/2022] Open
Abstract
Background: Clinical and pre-clinical studies indicate a reduction in seizure frequency as well as a decrease in susceptibility to subsequently evoked seizures after physical exercise programs. In contrast to the influence of exercise after epilepsy previously established, various studies have been conducted attempting to investigate whether physical activity reduces brain susceptibility to seizures or prevents epilepsy. We report a systematic review and meta-analysis of different animal models that addressed the impact of previous physical exercise programs to reduce seizure susceptibility. Methods: We included animal model (rats and mice) studies before brain insult that reported physical exercise programs compared with other interventions (sham, control, or naïve). We excluded studies that investigated animal models after brain insult, associated with supplement nutrition or drugs, that did not address epilepsy or seizure susceptibility, ex vivo studies, in vitro studies, studies in humans, or in silico studies. Electronic searches were performed in the MEDLINE (PubMed), Web of Science (WOS), Scopus, PsycINFO, Scientific Electronic Library Online (SciELO) databases, and gray literature, without restrictions to the year or language of publication. We used SYRCLE's risk of bias tool and CAMARADES checklist for study quality. We performed a synthesis of results for different types of exercise and susceptibility to seizures by random-effects meta-analysis. Results: Fifteen studies were included in the final analysis (543 animals), 13 of them used male animals, and Wistar rats were the most commonly studied species used in the studies (355 animals). The chemoconvulsants used in the selected studies were pentylenetetrazol, penicillin, kainic acid, pilocarpine, and homocysteine. We assessed the impact of study design characteristics and the reporting of mitigations to reduce the risk of bias. We calculated a standardized mean difference effect size for each comparison and performed a random-effects meta-analysis. The meta-analysis included behavioral analysis (latency to seizure onset, n = 6 and intensity of motor signals, n = 3) and electrophysiological analysis (spikes/min, n = 4, and amplitude, n = 6). The overall effect size observed in physical exercise compared to controls for latency to seizure onset was −130.98 [95% CI: −203.47, −58.49] (seconds) and the intensity of motor signals was −0.40 [95% CI: −1.19, 0.40] (on a scale from 0 to 5). The largest effects were observed in electrophysiological analysis for spikes/min with −26.96 [95% CI: −39.56, −14.36], and for spike amplitude (μV) with −282.64 [95% CI: −466.81, −98.47]. Discussion:Limitations of evidence. A higher number of animal models should be employed for analyzing the influence of exerciseon seizure susceptibility. The high heterogeneity in our meta-analysis is attributable to various factors, including the number of animals used in each study and the limited number of similar studies. Interpretation. Studies selected in this systematic review and meta-analysis suggest that previous physical exercise programs can reduce some of the main features related to seizure susceptibility [latency seizure onset, spikes/min, and spike amplitude (μV)] induced by the administration of different chemoconvulsants. Systematic Review Registration: PROSPERO, identifier CRD42021251949; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=251949.
Collapse
Affiliation(s)
- Ricardo Mario Arida
- Department of Physiology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | - Jean Faber
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | | |
Collapse
|
72
|
Martínez-Aguirre C, Cinar R, Rocha L. Targeting Endocannabinoid System in Epilepsy: For Good or for Bad. Neuroscience 2021; 482:172-185. [PMID: 34923038 DOI: 10.1016/j.neuroscience.2021.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
Epilepsy is a neurological disorder with a high prevalence worldwide. Several studies carried out during the last decades indicate that the administration of cannabinoids as well as the activation of the endocannabinoid system (ECS) represent a therapeutic strategy to control epilepsy. However, there are controversial studies indicating that activation of ECS results in cell damage, inflammation and neurotoxicity, conditions that facilitate the seizure activity. The present review is focused to present findings supporting this issue. According to the current discrepancies, it is relevant to elucidate the different effects induced by the activation of ECS and determine the conditions under which it facilitates the seizure activity.
Collapse
Affiliation(s)
| | - Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Rockville, USA
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies, Mexico City, Mexico.
| |
Collapse
|
73
|
The Endocannabinoid System in Glial Cells and Their Profitable Interactions to Treat Epilepsy: Evidence from Animal Models. Int J Mol Sci 2021; 22:ijms222413231. [PMID: 34948035 PMCID: PMC8709154 DOI: 10.3390/ijms222413231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is one of the most common neurological conditions. Yearly, five million people are diagnosed with epileptic-related disorders. The neuroprotective and therapeutic effect of (endo)cannabinoid compounds has been extensively investigated in several models of epilepsy. Therefore, the study of specific cell-type-dependent mechanisms underlying cannabinoid effects is crucial to understanding epileptic disorders. It is estimated that about 100 billion neurons and a roughly equal number of glial cells co-exist in the human brain. The glial population is in charge of neuronal viability, and therefore, their participation in brain pathophysiology is crucial. Furthermore, glial malfunctioning occurs in a wide range of neurological disorders. However, little is known about the impact of the endocannabinoid system (ECS) regulation over glial cells, even less in pathological conditions such as epilepsy. In this review, we aim to compile the existing knowledge on the role of the ECS in different cell types, with a particular emphasis on glial cells and their impact on epilepsy. Thus, we propose that glial cells could be a novel target for cannabinoid agents for treating the etiology of epilepsy and managing seizure-like disorders.
Collapse
|
74
|
|
75
|
Huang SS, Sheng YC, Jiang YY, Liu N, Lin MM, Wu JC, Liang ZQ, Qin ZH, Wang Y. TIGAR plays neuroprotective roles in KA-induced excitotoxicity through reducing neuroinflammation and improving mitochondrial function. Neurochem Int 2021; 152:105244. [PMID: 34826530 DOI: 10.1016/j.neuint.2021.105244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/30/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023]
Abstract
Excitotoxicity refers to the ability of excessive extracellular excitatory amino acids to damage neurons via receptor activation. It is a crucial pathogenetic process in neurodegenerative diseases. TP53 is confirmed to be involved in excitotoxicity. It is demonstrated that TP53 induced glycolysis and apoptotic regulator (TIGAR)-regulated metabolic pathway can protect against neuronal injury. However, the role of TIGAR in excitotoxicity and specific mechanisms is still unknown. In this study, an in vivo excitotoxicity model was constructed via stereotypical kainic acid (KA) injection into the striatum of mice. KA reduced TIGAR expression levels, neuroinflammatory responses and mitochondrial dysfunction. TIGAR overexpression could reverse KA-induced neuronal injury by reducing neuroinflammation and improving mitochondrial function, thereby exerting neuroprotective effects. Therefore, this study could provide a potential therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Si Huang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Chao Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Yue Jiang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Na Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Miao-Miao Lin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhong-Qin Liang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
76
|
Ahmed H, Khan MA, Ali Zaidi SA, Muhammad S. In Silico and In Vivo: Evaluating the Therapeutic Potential of Kaempferol, Quercetin, and Catechin to Treat Chronic Epilepsy in a Rat Model. Front Bioeng Biotechnol 2021; 9:754952. [PMID: 34805114 PMCID: PMC8599161 DOI: 10.3389/fbioe.2021.754952] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Recently, alternative therapies are gaining popularity in the treatment of epilepsy. The present study aimed to find out the antiepileptic potential of quercetin, catechin, and kaempferol. In vivo and in silico experiments were conducted to investigate their therapeutic potential. 25 mg/kg/day of pentylenetetrazole was administered for 4 weeks after epilepsy was induced in the rats; this was followed by the behavioral studies and histological analysis of rat brain slices. Binding affinities of kaempferol, quercetin, and catechin were assessed by performing in silico studies. Kaempferol, quercetin, and catechin were found to have the highest binding affinity with the synaptic vesicle 2A (SV2A) protein, comparable to standard levetiracetam (LEV). The mRNA levels of SV2A, as well as the expression of TNF, IL 6, IL 1 beta, NFkB, IL 1Ra, IL 4, and IL 10, were investigated using qPCR. Our results indicate for the first time that SV2A is also a transporter of understudied phytoflavonoids, due to which a significant improvement was observed in epileptic parameters. The mRNA levels of SV2A were found to be significantly elevated in the PF-treated rats when compared with those of the control rats with epilepsy. Additionally, downregulation of the pro-inflammatory cytokines and upregulation of the anti-inflammatory cytokines were also noted in the PF-treated groups. It is concluded that kaempferol, quercetin, and catechin can effectively decrease the epileptic seizures in our chronic epilepsy rat model to a level that is comparable to the antiepileptic effects induced by levetiracetam drug.
Collapse
Affiliation(s)
- Hammad Ahmed
- Faculty of Pharmacy, The University of Lahore, Defence Road Campus, Lahore, Pakistan.,Imran Idrees College of Pharmacy, Sialkot, Pakistan
| | | | | | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany.,Department of Neurosurgery, University of Helsinki and University Hospital, Helsinki, Finland
| |
Collapse
|
77
|
Torquato P, Giusepponi D, Bartolini D, Barola C, Marinelli R, Sebastiani B, Galarini R, Galli F. Pre-analytical monitoring and protection of oxidizable lipids in human plasma (vitamin E and ω-3 and ω-6 fatty acids): An update for redox-lipidomics methods. Free Radic Biol Med 2021; 176:142-148. [PMID: 34562608 DOI: 10.1016/j.freeradbiomed.2021.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022]
Abstract
Sample manipulation for storage and storage itself, interfere with the stability of labile lipids in human plasma, including vitamin E (α-tocopherol), polyunsaturated fatty acids (PUFAs), and their enzymatic and free radical-derived oxidation metabolites. This remains a main limit of lipidomics studies that often lack of sufficient standardization and validation at the pre-analytical level. In order to characterize the stability of these lipids in human plasma and to develop a standardized pre-analytical protocol for lipidomics methods, the oxidation metabolites of α-tocopherol, the free form of ω3 and ω6 PUFAs, and some arachidonic acid (AA)-derived eicosanoids were investigated in human plasma during storage at different freezing temperatures. The effect of a protection/defense cocktail of antioxidants and lipoxygenase inhibitors (PD solution) on these lipid parameters was also evaluated. The temperature of storage markedly affected the formation of α-tocopheryl quinone (α-TQ), the main lipoperoxyl radical-derived oxidation metabolite of vitamin E, with the lowest production rate observed in samples stored at -80 °C or in liquid nitrogen. A similar effect of the storage temperature was observed for the free form of the ω-3 species eicosapentaenoic and docosahexaenoic acid, and for the ω-6 AA. Freezing samples at -20 °C resulted in a time-dependent formation of the pro-inflammatory eicosanoid LTB4. The PD solution prevents non-specific alterations of these lipid parameters in samples that are processed for direct analysis and protects from the temperature-dependent modifications of free PUFAs. Combining PD solution and preservation at -80 °C or in liquid nitrogen, resulted in levels of α-TQ and PUFAs that remained stable over 1 month and up to 8 months of storage, respectively. This method paper provides indications for the optimal processing and storage of human plasma utilized in lipidomics studies.
Collapse
Affiliation(s)
- Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy
| | - Danilo Giusepponi
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati", 06126, Perugia, Italy.
| | - Desirée Bartolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06126, Perugia, Italy; Dept. of Medicine and Surgery (Section of Human, Clinical and Forensic Anatomy), University of Perugia, 06126, Perugia, Italy.
| | - Carolina Barola
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati", 06126, Perugia, Italy.
| | - Rita Marinelli
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy.
| | - Bartolomeo Sebastiani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06126, Perugia, Italy.
| | - Roberta Galarini
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati", 06126, Perugia, Italy.
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy.
| |
Collapse
|
78
|
Tapia A, Giachello CN, Palomino-Schätzlein M, Baines RA, Galindo MI. Generation and Characterization of the Drosophila melanogaster paralytic Gene Knock-Out as a Model for Dravet Syndrome. Life (Basel) 2021; 11:life11111261. [PMID: 34833136 PMCID: PMC8619338 DOI: 10.3390/life11111261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Dravet syndrome is a severe rare epileptic disease caused by mutations in the SCN1A gene coding for the Nav1.1 protein, a voltage-gated sodium channel alpha subunit. We have made a knock-out of the paralytic gene, the single Drosophila melanogaster gene encoding this type of protein, by homologous recombination. These flies showed a heat-induced seizing phenotype, and sudden death in long term seizures. In addition to seizures, neuromuscular alterations were observed in climbing, flight, and walking tests. Moreover, they also manifested some cognitive alterations, such as anxiety and problems in learning. Electrophysiological analyses from larval motor neurons showed a decrease in cell capacitance and membrane excitability, while persistent sodium current increased. To detect alterations in metabolism, we performed an NMR metabolomic profiling of heads, which revealed higher levels in some amino acids, succinate, and lactate; and also an increase in the abundance of GABA, which is the main neurotransmitter implicated in Dravet syndrome. All these changes in the paralytic knock-out flies indicate that this is a good model for epilepsy and specifically for Dravet syndrome. This model could be a new tool to understand the pathophysiology of the disease and to find biomarkers, genetic modifiers and new treatments.
Collapse
Affiliation(s)
- Andrea Tapia
- Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (A.T.); (M.P.-S.)
| | - Carlo N. Giachello
- Manchester Academic Health Science Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (C.N.G.); (R.A.B.)
| | | | - Richard A. Baines
- Manchester Academic Health Science Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (C.N.G.); (R.A.B.)
| | - Máximo Ibo Galindo
- Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (A.T.); (M.P.-S.)
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
- UPV-CIPF Joint Unit Disease Mechanisms and Nanomedicine, 46012 Valencia, Spain
- Correspondence:
| |
Collapse
|
79
|
Zhao H, Li S, He L, Tang F, Han X, Deng W, Lin Z, Huang R, Li Z. Ameliorating Effect of Umbilical Cord Mesenchymal Stem Cells in a Human Induced Pluripotent Stem Cell Model of Dravet Syndrome. Mol Neurobiol 2021; 59:748-761. [PMID: 34766239 DOI: 10.1007/s12035-021-02633-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023]
Abstract
Dravet syndrome (DS) is a form of severe childhood-onset refractory epilepsy typically caused by a heterozygous loss-of-function mutation. DS patient-derived induced pluripotent stem cells (iPSCs) are appropriate human cells for exploring disease mechanisms and testing new therapeutic strategies in vitro. Repeated spontaneous seizures can cause neuroinflammatory reactions and oxidative stress, resulting in neuronal toxicity, neuronal dysfunction, blood-brain barrier disruption, and hippocampal inflammation. Antiepileptic drug therapy does not delay the development of chronic epilepsy. The application of mesenchymal stem cells (MSCs) is one therapeutic strategy for thwarting epilepsy development. This study evaluated the effects of human umbilical cord mesenchymal stem cell-conditioned medium (HUMSC-CM) in a new in vitro model of neurons differentiated from DS patient-derived iPSCs. In the presence of HUMSC-CM, increases in superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2), glutathione peroxidase (GPX), and glutathione (GSH) levels were found to contribute to a reduction in reactive oxygen species (ROS) levels. In parallel, inflammation was rescued in DS patient-derived neuronal cells via increased expression of anti-inflammatory cytokines (TGF-β, IL-6, and IL-10) and significant downregulation of tumor necrosis factor-α and interleukin-1β expression. The intracellular calcium concentration ([Ca2+]i) and malondialdehyde (MDA) and ROS levels were decreased in DS patient-derived cells. In addition, action potential (AP) firing ability was enhanced by HUMSC-CM. In conclusion, HUMSC-CM can effectively eliminate ROS, affect migration and neurogenesis, and promote neurons to enter a highly functional state. Therefore, HUMSC-CM is a promising therapeutic strategy for the clinical treatment of refractory epilepsy such as DS.
Collapse
Affiliation(s)
- Huifang Zhao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Shuai Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lang He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Tang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaobo Han
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangzhou Medical University, Guangzhou, 511436, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiyue Deng
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zuoxian Lin
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Rongqi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhiyuan Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China.
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Guangzhou Medical University, Guangzhou, 511436, China.
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
80
|
The Protective Role of E-64d in Hippocampal Excitotoxic Neuronal Injury Induced by Glutamate in HT22 Hippocampal Neuronal Cells. Neural Plast 2021; 2021:7174287. [PMID: 34721570 PMCID: PMC8550833 DOI: 10.1155/2021/7174287] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 12/25/2022] Open
Abstract
Epilepsy is the most common childhood neurologic disorder. Status epilepticus (SE), which refers to continuous epileptic seizures, occurs more frequently in children than in adults, and approximately 40–50% of all cases occur in children under 2 years of age. Conventional antiepileptic drugs currently used in clinical practice have a number of adverse side effects. Drug-resistant epilepsy (DRE) can progressively develop in children with persistent SE, necessitating the development of novel therapeutic drugs. During SE, the persistent activation of neurons leads to decreased glutamate clearance with corresponding glutamate accumulation in the synaptic extracellular space, increasing the chance of neuronal excitotoxicity. Our previous study demonstrated that after developmental seizures in rats, E-64d exerts a neuroprotective effect on the seizure-induced brain damage by modulating lipid metabolism enzymes, especially ApoE and ApoJ/clusterin. In this study, we investigated the impact and mechanisms of E-64d administration on neuronal excitotoxicity. To test our hypothesis that E-64d confers neuroprotective effects by regulating autophagy and mitochondrial pathway activity, we simulated neuronal excitotoxicity in vitro using an immortalized hippocampal neuron cell line (HT22). We found that E-64d improved cell viability while reducing oxidative stress and neuronal apoptosis. In addition, E-64d treatment regulated mitochondrial pathway activity and inhibited chaperone-mediated autophagy in HT22 cells. Our findings indicate that E-64d may alleviate glutamate-induced damage via regulation of mitochondrial fission and apoptosis, as well as inhibition of chaperone-mediated autophagy. Thus, E-64d may be a promising therapeutic treatment for hippocampal injury associated with SE.
Collapse
|
81
|
Zhu J, Li L, Ding J, Huang J, Shao A, Tang B. The Role of Formyl Peptide Receptors in Neurological Diseases via Regulating Inflammation. Front Cell Neurosci 2021; 15:753832. [PMID: 34650406 PMCID: PMC8510628 DOI: 10.3389/fncel.2021.753832] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a group of G protein-coupled cell surface receptors that play important roles in host defense and inflammation. Owing to the ubiquitous expression of FPRs throughout different cell types and since they interact with structurally diverse chemotactic agonists, they have a dual function in inflammatory processes, depending on binding with different ligands so that accelerate or inhibit key intracellular kinase-based regulatory pathways. Neuroinflammation is closely associated with the pathogenesis of neurodegenerative diseases, neurogenic tumors and cerebrovascular diseases. From recent studies, it is clear that FPRs are important biomarkers for neurological diseases as they regulate inflammatory responses by monitoring glial activation, accelerating neural differentiation, regulating angiogenesis, and controlling blood brain barrier (BBB) permeability, thereby affecting neurological disease progression. Given the complex mechanisms of neurological diseases and the difficulty of healing, we are eager to find new and effective therapeutic targets. Here, we review recent research about various mechanisms of the effects generated after FPR binding to different ligands, role of FPRs in neuroinflammation as well as the development and prognosis of neurological diseases. We summarize that the FPR family has dual inflammatory functional properties in central nervous system. Emphasizing that FPR2 acts as a key molecule that mediates the active resolution of inflammation, which binds with corresponding receptors to reduce the expression and activation of pro-inflammatory composition, govern the transport of immune cells to inflammatory tissues, and restore the integrity of the BBB. Concurrently, FPR1 is essentially related to angiogenesis, cell proliferation and neurogenesis. Thus, treatment with FPRs-modulation may be effective for neurological diseases.
Collapse
Affiliation(s)
- Jiahui Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingfei Li
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiao Ding
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinyu Huang
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Tang
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
82
|
Holton KF. Micronutrients May Be a Unique Weapon Against the Neurotoxic Triad of Excitotoxicity, Oxidative Stress and Neuroinflammation: A Perspective. Front Neurosci 2021; 15:726457. [PMID: 34630015 PMCID: PMC8492967 DOI: 10.3389/fnins.2021.726457] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Excitotoxicity has been implicated in many neurological disorders and is a leading cause of oxidative stress and neuroinflammation in the nervous system. Most of the research to date has focused on each of these conditions individually; however, excitotoxicity, oxidative stress, and neuroinflammation have the ability to influence one another in a self-sustaining manner, thus functioning as a "neurotoxic triad." This perspective article re-introduces the concept of the neurotoxic triad and reviews how specific dietary micronutrients have been shown to protect against not only oxidative stress, but also excitotoxicity and neuroinflammation. Future dietary interventions for neurological disorders could focus on the effects on all three aspects of the neurotoxic triad.
Collapse
Affiliation(s)
- Kathleen F Holton
- Nutritional Neuroscience Lab, Department of Health Studies, Center for Neuroscience and Behavior, American University, Washington, DC, United States
| |
Collapse
|
83
|
Carveol Attenuates Seizure Severity and Neuroinflammation in Pentylenetetrazole-Kindled Epileptic Rats by Regulating the Nrf2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9966663. [PMID: 34422216 PMCID: PMC8376446 DOI: 10.1155/2021/9966663] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022]
Abstract
Epilepsy is a neurodegenerative brain disorder characterized by recurrent seizure attacks. Numerous studies have suggested a strong correlation between oxidative stress and neuroinflammation in several neurodegenerative disorders including epilepsy. This study is aimed at investigating the neuroprotective effects of the natural compound carveol against pentylenetetrazole- (PTZ-) induced kindling and seizure model. Two different doses of carveol (10 mg/kg and 20 mg/kg) were administered to male rats to determine the effects and the effective dose of carveol and to further demonstrate the mechanism of action of nuclear factor E2-related factor (Nrf2) in PTZ-induced kindling model. Our results demonstrated reduced levels of innate antioxidants such as superoxide dismutase (SOD), catalase, glutathione-S-transferase (GST), and glutathione (GSH), associated with elevated lipid peroxidation (LPO) and inflammatory cytokines level such as tumor necrosis factor-alpha (TNF-α), and mediators like cyclooxygenase (COX-2) and nuclear factor kappa B (NFκB). These detrimental effects exacerbated oxidative stress and provoked a marked neuronal alteration in the cortex and hippocampus of PTZ-intoxicated animals that were associated with upregulated Nrf2 gene expression. Furthermore, carveol treatment positively modulated the antioxidant gene Nrf2 and its downstream target HO-1. To further investigate the role of Nrf2, an inhibitor of Nrf2 called all-trans retinoic acid (ATRA) was used, which further exacerbated PTZ toxicity. Moreover, carveol treatment induced cholinergic system activation by mitigating acetylcholinesterase level which is further linked to attenuated neuroinflammatory cascade. The extent of blood-brain barrier disruption was evaluated based on vascular endothelial growth factor (VEGF) expression. Taken together, our findings suggest that carveol acts as an Nrf2 activator and therefore induces downstream antioxidants and mitigates inflammatory insults through multiple pathways. This eventually alleviates PTZ-induced neuroinflammation and neurodegeneration.
Collapse
|
84
|
Kumar S, Kumar P. The Beneficial Effect of Rice Bran Extract Against Rotenone-Induced Experimental Parkinson's Disease in Rats. Curr Mol Pharmacol 2021; 14:428-438. [PMID: 33573588 DOI: 10.2174/1874467214666210126113324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Neurodegenerative diseases have become an increasing cause of various disabilities worldwide, followed by aging, including Parkinson's disease (PD). Parkinson's disease is a degenerative brain disorder distinguished by growing motor & non-motor failure due to the degeneration of medium-sized spiked neurons in the striatum region. Rotenone is often employed to originate the animal model of PD. It is a powerful blocker of mitochondrial complex-I, mitochondrial electron transport chain that reliably produces Parkinsonism-like symptoms in rats. Rice bran (RB) is very rich in polyunsaturated fatty acids (PUFA) and nutritionally beneficial compounds, such as γ-oryzanol, tocopherols, and tocotrienols and sterols are believed to have favorable outcomes on oxidative stress & mitochondrial function. OBJECTIVE The present study has been designed to explore RB extract's effect against rotenone-induced neurotoxicity in rats. METHODS In the present study, Rotenone (2 mg/kg, s.c) was administered systemically for 28 days. The hexane extract of RB was prepared using Soxhlation. Hexane extract (250 & 500 mg/kg) was administered per oral for 28 days in rotenone-treated groups. Behavioral parameters (grip strength, motor coordination, locomotion, and catalepsy) were conducted on the 7th, 14th, 21st, and 28th day. Animals were sacrificed on the 29th day for biochemical estimation in the striatum and cortex. RESULTS This study demonstrates significant alteration in behavioral parameters, oxidative burden (increased lipid peroxidation, nitrite concentration, and decreased glutathione, catalase, SOD) in rotenone-treated animals. Administration of hexane extract of RB prevented the behavioral, biochemical alterations induced by rotenone. The current research has been sketched to inspect RB extract's effect against rotenone-developed neurotoxicity in rats. CONCLUSION The findings support that PD is associated with impairments in motor activity. The results also suggest that the nutraceutical rice bran that contains γ-oryzanol, Vitamin-E, ferulic acid etc., may underlie the adjuvant susceptibility towards rotenone-induced PD in experimental rats.
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Puneet Kumar
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| |
Collapse
|
85
|
Petrillo S, Pietrafusa N, Trivisano M, Calabrese C, Saura F, Gallo MG, Bertini ES, Vigevano F, Specchio N, Piemonte F. Imbalance of Systemic Redox Biomarkers in Children with Epilepsy: Role of Ferroptosis. Antioxidants (Basel) 2021; 10:antiox10081267. [PMID: 34439515 PMCID: PMC8389337 DOI: 10.3390/antiox10081267] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022] Open
Abstract
To assess if ferroptosis, a new type of programmed cell death accompanied by iron accumulation, lipid peroxidation, and glutathione depletion, occurs in children with epilepsy, and in order to identify a panel of biomarkers useful for patient stratification and innovative-targeted therapies, we measured ferroptosis biomarkers in blood from 83 unrelated children with a clinical diagnosis of epilepsy and 44 age-matched controls. We found a marked dysregulation of three ferroptosis key markers: a consistent increase of 4-hydroxy-2-nonenal (4-HNE), the main by-product of lipid peroxidation, a significant decrease of glutathione (GSH) levels, and a partial inactivation of the enzyme glutathione peroxidase 4 (GPX4), the mediator of lipid peroxides detoxification. Furthermore, we found a significant increase of NAPDH oxidase 2 (NOX2) in the blood of children, supporting this enzyme as a primary source of reactive oxygen species (ROS) in epilepsy. Additionally, since the nuclear factor erythroid 2-related factor 2 (NRF2) induction protects the brain from epileptic seizure damage, we also evaluated the NRF2 expression in the blood of children. The antioxidant and anti-inflammatory transcription factor was activated in patients, although not enough to re-establish a correct redox homeostasis for counteracting ferroptosis. Ferroptosis-mediated oxidative damage has been proposed as an emergent mechanism underlying the pathogenesis of epilepsy. Overall, our study confirms a crucial role for ferroptosis in epilepsy, leading to the identification of a panel of biomarkers useful to find new therapeutic targets. Developing innovative drugs, which act by inhibiting the ferroptosis signaling axis, may represent a promising strategy for new anti-seizure medications.
Collapse
Affiliation(s)
- Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy; (S.P.); (M.G.G.); (E.S.B.)
| | - Nicola Pietrafusa
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Piazza S. Onofrio 4, 00165 Rome, Italy; (N.P.); (M.T.); (C.C.)
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Piazza S. Onofrio 4, 00165 Rome, Italy; (N.P.); (M.T.); (C.C.)
| | - Costanza Calabrese
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Piazza S. Onofrio 4, 00165 Rome, Italy; (N.P.); (M.T.); (C.C.)
| | - Francesca Saura
- Department of Laboratory Medicine, Children’s Hospital Bambino Gesù, Piazza S. Onofrio 4, 00165 Rome, Italy;
| | - Maria Giovanna Gallo
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy; (S.P.); (M.G.G.); (E.S.B.)
| | - Enrico Silvio Bertini
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy; (S.P.); (M.G.G.); (E.S.B.)
| | - Federico Vigevano
- Department of Neuroscience, Bambino Gesu Children’s Hospital, IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Piazza S. Onforio 4, 00165 Rome, Italy; (F.V.); (N.S.)
| | - Nicola Specchio
- Department of Neuroscience, Bambino Gesu Children’s Hospital, IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Piazza S. Onforio 4, 00165 Rome, Italy; (F.V.); (N.S.)
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy; (S.P.); (M.G.G.); (E.S.B.)
- Correspondence: ; Tel.: +39-06-6859-2102
| |
Collapse
|
86
|
Hwang Y, Kim HC, Shin EJ. Repeated exposure to microcystin-leucine-arginine potentiates excitotoxicity induced by a low dose of kainate. Toxicology 2021; 460:152887. [PMID: 34352349 DOI: 10.1016/j.tox.2021.152887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/15/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Microcystin-leucine-arginine (MLCR) is a cyanobacterial toxin, and has been demonstrated to cause neurotoxicity. In addition, MCLR has been identified as an inhibitor of protein phosphatase (PP)1 and PP2A, which are known to regulate the phosphorylation of various molecules related to synaptic excitability. Thus, in the present study, we examined whether MCLR exposure affects seizures induced by a low dose of kainic acid (KA; 0.05 μg, i.c.v.) administration. KA-induced seizure occurrence and seizure score significantly increased after repeated exposure to MCLR (2.5 or 5.0 μg/kg, i.p., once a day for 10 days), but not after acute MCLR exposure (2.5 or 5.0 μg/kg, i.p., 2 h and 30 min prior to KA administration), and hippocampal neuronal loss was consistently facilitated by repeated exposure to MCLR. In addition, repeated MCLR significantly elevated the membrane expression of kainate receptor GluK2 subunits, p-pan-protein kinase C (PKC), and p-extracellular signal-related kinase (ERK) at 1 h after KA. However, KA-induced membrane expression of Ca2+/calmodulin-dependent kinase II (CaMKII) was significantly reduced by repeated MCLR exposure. Consistent with the enhanced seizures and neurodegeneration, MCLR exposure significantly potentiated KA-induced oxidative stress and microglial activation, which was accompanied by increased expression of p-ERK and p-PKCδ in the hippocampus. The combined results suggest that repeated MCLR exposure potentiates KA-induced excitotoxicity in the hippocampus by increasing membrane GluK2 expression and enhancing oxidative stress and neuroinflammation through the modulation of p-CaMKII, p-PKC, and p-ERK.
Collapse
Affiliation(s)
- Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
87
|
Selivanov VA, Zagubnaya OA, Nartsissov YR, Cascante M. Unveiling a key role of oxaloacetate-glutamate interaction in regulation of respiration and ROS generation in nonsynaptic brain mitochondria using a kinetic model. PLoS One 2021; 16:e0255164. [PMID: 34343196 PMCID: PMC8330910 DOI: 10.1371/journal.pone.0255164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022] Open
Abstract
Glutamate plays diverse roles in neuronal cells, affecting cell energetics and reactive oxygen species (ROS) generation. These roles are especially vital for neuronal cells, which deal with high amounts of glutamate as a neurotransmitter. Our analysis explored neuronal glutamate implication in cellular energy metabolism and ROS generation, using a kinetic model that simulates electron transport details in respiratory complexes, linked ROS generation and metabolic reactions. The analysis focused on the fact that glutamate attenuates complex II inhibition by oxaloacetate, stimulating the latter's transformation into aspartate. Such a mechanism of complex II activation by glutamate could cause almost complete reduction of ubiquinone and deficiency of oxidized form (Q), which closes the main stream of electron transport and opens a way to massive ROS generating transfer in complex III from semiquinone radicals to molecular oxygen. In this way, under low workload, glutamate triggers the respiratory chain (RC) into a different steady state characterized by high ROS generation rate. The observed stepwise dependence of ROS generation on glutamate concentration experimentally validated this prediction. However, glutamate's attenuation of oxaloacetate's inhibition accelerates electron transport under high workload. Glutamate-oxaloacetate interaction in complex II regulation underlies the observed effects of uncouplers and inhibitors and acceleration of Ca2+ uptake. Thus, this theoretical analysis uncovered the previously unknown roles of oxaloacetate as a regulator of ROS generation and glutamate as a modifier of this regulation. The model predicted that this mechanism of complex II activation by glutamate might be operative in situ and responsible for excitotoxicity. Spatial-time gradients of synthesized hydrogen peroxide concentration, calculated in the reaction-diffusion model with convection under a non-uniform local approximation of nervous tissue, have shown that overproduction of H2O2 in a cell causes excess of its level in neighbor cells.
Collapse
Affiliation(s)
- Vitaly A. Selivanov
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES- ELIXIR), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Olga A. Zagubnaya
- Department of Mathematical Modeling and Statistical Analysis, Institute of Cytochemistry and Molecular Pharmacology, Moscow, Russia
| | - Yaroslav R. Nartsissov
- Department of Mathematical Modeling and Statistical Analysis, Institute of Cytochemistry and Molecular Pharmacology, Moscow, Russia
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES- ELIXIR), Institute of Health Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
88
|
Santos VR, Melo IS, Pacheco ALD, Castro OWD. Life and death in the hippocampus: What's bad? Epilepsy Behav 2021; 121:106595. [PMID: 31759972 DOI: 10.1016/j.yebeh.2019.106595] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/13/2023]
Abstract
The hippocampal formation is crucial for the generation and regulation of several brain functions, including memory and learning processes; however, it is vulnerable to neurological disorders, such as epilepsy. Temporal lobe epilepsy (TLE), the most common type of epilepsy, changes the hippocampal circuitry and excitability, under the contribution of both neuronal degeneration and abnormal neurogenesis. Classically, neurodegeneration affects sensitive areas of the hippocampus, such as dentate gyrus (DG) hilus, as well as specific fields of the Ammon's horn, CA3, and CA1. In addition, the proliferation, migration, and abnormal integration of newly generated hippocampal granular cells (GCs) into the brain characterize TLE neurogenesis. Robust studies over the years have intensely discussed the effects of death and life in the hippocampus, though there are still questions to be answered about their possible benefits and risks. Here, we review the impacts of death and life in the hippocampus, discussing its influence on TLE, providing new perspectives or insights for the implementation of new possible therapeutic targets. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Victor Rodrigues Santos
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Igor Santana Melo
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil
| | | | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil.
| |
Collapse
|
89
|
Dash R, Mitra S, Ali MC, Oktaviani DF, Hannan MA, Choi SM, Moon IS. Phytosterols: Targeting Neuroinflammation in Neurodegeneration. Curr Pharm Des 2021; 27:383-401. [PMID: 32600224 DOI: 10.2174/1381612826666200628022812] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/02/2020] [Indexed: 11/22/2022]
Abstract
Plant-derived sterols, phytosterols, are well known for their cholesterol-lowering activity in serum and their anti-inflammatory activities. Recently, phytosterols have received considerable attention due to their beneficial effects on various non-communicable diseases, and recommended use as daily dietary components. The signaling pathways mediated in the brain by phytosterols have been evaluated, but little is known about their effects on neuroinflammation, and no clinical studies have been undertaken on phytosterols of interest. In this review, we discuss the beneficial roles of phytosterols, including their attenuating effects on inflammation, blood cholesterol levels, and hallmarks of the disease, and their regulatory effects on neuroinflammatory disease pathways. Despite recent advancements made in phytosterol pharmacology, some critical questions remain unanswered. Therefore, we have tried to highlight the potential of phytosterols as viable therapeutics against neuroinflammation and to direct future research with respect to clinical applications.
Collapse
Affiliation(s)
- Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Sarmistha Mitra
- Plasma Bioscience Research Center, Plasma Bio-display, Kwangwoon University, Seoul-01897, Korea
| | - Md Chayan Ali
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Diyah Fatimah Oktaviani
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Md Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| | - Sung Min Choi
- Department of Pediatrics, Dongguk University College of Medicine, Gyeongju-38066, Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea
| |
Collapse
|
90
|
Contreras-García IJ, Gómez-Lira G, Phillips-Farfán BV, Pichardo-Macías LA, García-Cruz ME, Chávez-Pacheco JL, Mendoza-Torreblanca JG. Synaptic Vesicle Protein 2A Expression in Glutamatergic Terminals Is Associated with the Response to Levetiracetam Treatment. Brain Sci 2021; 11:brainsci11050531. [PMID: 33922424 PMCID: PMC8145097 DOI: 10.3390/brainsci11050531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/05/2022] Open
Abstract
Synaptic vesicle protein 2A (SV2A), the target of the antiepileptic drug levetiracetam (LEV), is expressed ubiquitously in all synaptic terminals. Its levels decrease in patients and animal models of epilepsy. Thus, changes in SV2A expression could be a critical factor in the response to LEV. Epilepsy is characterized by an imbalance between excitation and inhibition, hence SV2A levels in particular terminals could also influence the LEV response. SV2A expression was analyzed in the epileptic hippocampus of rats which responded or not to LEV, to clarify if changes in SV2A alone or together with glutamatergic or GABAergic markers may predict LEV resistance. Wistar rats were administered saline (control) or pilocarpine to induce epilepsy. These groups were subdivided into untreated or LEV-treated groups. All epileptic rats were video-monitored to assess their number of seizures. Epileptic rats with an important seizure reduction (>50%) were classified as responders. SV2A, vesicular γ-aminobutyric acid transporter and vesicular glutamate transporter (VGLUT) expression were assessed by immunostaining. SV2A expression was not modified during epilepsy. However, responders showed ≈55% SV2A-VGLUT co-expression in comparison with the non-responder group (≈40%). Thus, SV2A expression in glutamatergic terminals may be important for the response to LEV treatment.
Collapse
Affiliation(s)
- Itzel Jatziri Contreras-García
- Área de Neurociencias, Biología de la Reproducción, Unidad Iztapalapa, Universidad Autónoma Metropolitana, Ciudad de México 09340, Mexico;
| | - Gisela Gómez-Lira
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 14330, Mexico;
| | - Bryan Víctor Phillips-Farfán
- Laboratorio de Nutrición Experimental, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Ciudad de México 07738, Mexico;
| | - Mercedes Edna García-Cruz
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Juan Luis Chávez-Pacheco
- Laboratorio de Farmacología, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Julieta G. Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: ; Tel.: +52-55-840900 (ext. 1425) or +52-55-21836345
| |
Collapse
|
91
|
Potential Effects of Melatonin and Micronutrients on Mitochondrial Dysfunction during a Cytokine Storm Typical of Oxidative/Inflammatory Diseases. Diseases 2021; 9:diseases9020030. [PMID: 33919780 PMCID: PMC8167770 DOI: 10.3390/diseases9020030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Exaggerated oxidative stress and hyper-inflammation are essential features of oxidative/inflammatory diseases. Simultaneously, both processes may be the cause or consequence of mitochondrial dysfunction, thus establishing a vicious cycle among these three factors. However, several natural substances, including melatonin and micronutrients, may prevent or attenuate mitochondrial damage and may preserve an optimal state of health by managing the general oxidative and inflammatory status. This review aims to describe the crucial role of mitochondria in the development and progression of multiple diseases as well as the close relationship among mitochondrial dysfunction, oxidative stress, and cytokine storm. Likewise, it attempts to summarize the main findings related to the powerful effects of melatonin and some micronutrients (vitamins and minerals), which may be useful (alone or in combination) as therapeutic agents in the treatment of several examples of oxidative/inflammatory pathologies, including sepsis, as well as cardiovascular, renal, neurodegenerative, and metabolic disorders.
Collapse
|
92
|
Decoding signaling pathways involved in prolactin-induced neuroprotection: A review. Front Neuroendocrinol 2021; 61:100913. [PMID: 33766566 DOI: 10.1016/j.yfrne.2021.100913] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 11/23/2022]
Abstract
It has been well recognized that prolactin (PRL), a pleiotropic hormone, has many functions in the brain, such as maternal behavior, neurogenesis, and neuronal plasticity, among others. Recently, it has been reported to have a significant role in neuroprotection against excitotoxicity. Glutamate excitotoxicity is a common alteration in many neurological and neurodegenerative diseases, leading to neuronal death. In this sense, several efforts have been made to decrease the progression of these pathologies. Despite various reports of PRL's neuroprotective effect against excitotoxicity, the signaling pathways that underlie this mechanism remain unclear. This review aims to describe the most recent and relevant studies on the molecular signaling pathways, particularly, PI3K/AKT, NF-κB, and JAK2/STAT5, which are currently under investigation and might be implicated in the molecular mechanisms that explain the PRL effects against excitotoxicity and neuroprotection. Remarkable neuroprotective effects of PRL might be useful in the treatment of some neurological diseases.
Collapse
|
93
|
Lin CH, Chou IC, Hong SY. Genetic factors and the risk of drug-resistant epilepsy in young children with epilepsy and neurodevelopment disability: A prospective study and updated meta-analysis. Medicine (Baltimore) 2021; 100:e25277. [PMID: 33761731 PMCID: PMC8049163 DOI: 10.1097/md.0000000000025277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/03/2021] [Indexed: 11/26/2022] Open
Abstract
Drug-resistant epilepsy (DRE) affects 7% to 20% of children with epilepsy. Although some risk factors for DRE have been identified, the results have not been consistent. Moreover, data regarding the risk factors for epilepsy and its seizure outcome in the first 2 years of life are limited.We analyzed data for children aged 0 to 2 years with epilepsy and neurodevelopmental disability from January, 2013, through December, 2017. These patients were followed up to compare the risk of DRE in patients with genetic defect (genetic group) with that without genetic defect (nongenetic group). Additionally, we conducted a meta-analysis to identify the pooled prevalence of genetic factors in children with DRE.A total of 96 patients were enrolled. A total of 68 patients were enrolled in the nongenetic group, whereas 28 patients were enrolled in the genetic group. The overall DRE risk in the genetic group was 6.5 times (95% confidence interval [CI], 2.15-19.6; p = 0.03) higher than that in the nongenetic group. Separately, a total of 1308 DRE patients were participated in the meta-analysis. The pooled prevalence of these patients with genetic factors was 22.8% (95% CI 17.4-29.3).The genetic defect plays a crucial role in the development of DRE in younger children with epilepsy and neurodevelopmental disability. The results can serve as a reference for further studies of epilepsy panel design and may also assist in the development of improved treatments and prevention strategies for DRE.
Collapse
Affiliation(s)
- Chien-Heng Lin
- Division of Pediatrics Pulmonology, China Medical University, Children's Hospital, Taichung, Taiwan
- Department of Biomedical Imaging and Radiological Science, College of Medicine, China Medical University
| | - I-Ching Chou
- Division of Pediatrics Neurology, China Medical University, Children's Hospital
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Syuan-Yu Hong
- Division of Pediatrics Neurology, China Medical University, Children's Hospital
| |
Collapse
|
94
|
de Melo IS, Dos Santos YMO, Pacheco ALD, Costa MA, de Oliveira Silva V, Freitas-Santos J, de Melo Bastos Cavalcante C, Silva-Filho RC, Leite ACR, Gitaí DGL, Duzzioni M, Sabino-Silva R, Borbely AU, de Castro OW. Role of Modulation of Hippocampal Glucose Following Pilocarpine-Induced Status Epilepticus. Mol Neurobiol 2021; 58:1217-1236. [PMID: 33123979 DOI: 10.1007/s12035-020-02173-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
Abstract
Status epilepticus (SE) is defined as continuous and self-sustaining seizures, which trigger hippocampal neurodegeneration, mitochondrial dysfunction, oxidative stress, and energy failure. During SE, the neurons become overexcited, increasing energy consumption. Glucose uptake is increased via the sodium glucose cotransporter 1 (SGLT1) in the hippocampus under epileptic conditions. In addition, modulation of glucose can prevent neuronal damage caused by SE. Here, we evaluated the effect of increased glucose availability in behavior of limbic seizures, memory dysfunction, neurodegeneration process, neuronal activity, and SGLT1 expression. Vehicle (VEH, saline 0.9%, 1 μL) or glucose (GLU; 1, 2 or 3 mM, 1 μL) were administered into hippocampus of male Wistar rats (Rattus norvegicus) before or after pilocarpine to induce SE. Behavioral analysis of seizures was performed for 90 min during SE. The memory and learning processes were analyzed by the inhibitory avoidance test. After 24 h of SE, neurodegeneration process, neuronal activity, and SGLT1 expression were evaluated in hippocampal and extrahippocampal regions. Modulation of hippocampal glucose did not protect memory dysfunction followed by SE. Our results showed that the administration of glucose after pilocarpine reduced the severity of seizures, as well as the number of limbic seizures. Similarly, glucose after SE reduced cell death and neuronal activity in hippocampus, subiculum, thalamus, amygdala, and cortical areas. Finally, glucose infusion elevated the SGLT1 expression in hippocampus. Taken together our data suggest that possibly the administration of intrahippocampal glucose protects brain in the earlier stage of epileptogenic processes via an important support of SGLT1.
Collapse
Affiliation(s)
- Igor Santana de Melo
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | | | - Amanda Larissa Dias Pacheco
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Maisa Araújo Costa
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Vanessa de Oliveira Silva
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Jucilene Freitas-Santos
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | | | - Reginaldo Correia Silva-Filho
- Bioenergetics Laboratory, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Ana Catarina Rezende Leite
- Bioenergetics Laboratory, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Daniel Góes Leite Gitaí
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Marcelo Duzzioni
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, MG, Brazil
| | - Alexandre Urban Borbely
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil.
| |
Collapse
|
95
|
Aghdash SN. Herbal Medicine in the Treatment of Epilepsy. Curr Drug Targets 2021; 22:356-367. [PMID: 33023444 DOI: 10.2174/1389450121999201001152221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 11/22/2022]
Abstract
Epilepsy is one of the most common disorders of the central nervous system. Although epilepsy is common worldwide, approximately 80% of epileptic patients live in the developing countries or those with low-middle income. Up until the second decade of the 20th century, epilepsy was treated mostly by traditional remedies. Today, antiepileptic drugs are used as a general treatment instead to prevent and control epileptic seizures. However, patient access to these drugs is hindered due to the healthcare systems of their countries and a number of other reasons, such as cultural, socio-demographic, and financial poverty. In addition, approximately 30-40%of epileptic patients suffer from refractory epilepsy, additionally, AEDs have adverse side-effects that can lead to treatment failure or reduce the patient's quality of life. Despite recent advances in the treatment of epilepsy, there is still a need for improving medical treatment with a particular focus on efficacy, safety, and accessibility. Since herbal medicines have been used for many centuries around the world for treating epilepsy, it is, therefore, plausible that a rigorous study on herbal medicine and phytochemical components within plants of various species and origin may lead to the discovery of novel AEDs. Nowadays, many medicinal plants used in different cultures and regions of the world have been identified. Most phytochemical components of these plants have been identified and, in some cases, their targets located. Therefore, it is possible that new, effective, and accessible anticonvulsants drugs can be obtained from a medicinal plant.
Collapse
Affiliation(s)
- Simin Namvar Aghdash
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
96
|
Sarahian N, Mohammadi MT, Darabi S, Faghihi N. Fenofibrate protects the neurovascular unit and ameliorates plasma corticosterone levels in pentylenetetrazole-induced kindling seizure in mice. Brain Res 2021; 1758:147343. [PMID: 33556377 DOI: 10.1016/j.brainres.2021.147343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/03/2021] [Accepted: 01/31/2021] [Indexed: 02/07/2023]
Abstract
Epileptic seizures are the most common neurological diseases that change the function of neurovascular unit at molecular levels accompanied by activation of a wide variety of neurodegenerative cascades. Based on the pleiotropic functions of peroxisome proliferator-activated receptor-alpha (PPARα), the current study evaluated the neuroprotective effects of fenofibrate (an effective PPARα agonist) on the brain injuries induced by pentylenetetrazole (PTZ)-induced kindling seizure. Adult male NMRI mice were randomly assigned into four groups (n = 14) as follows; control, untreated kindled mice (PTZ) and two fenofibrate-treated kindled groups. Repeated intraperitoneal injections of PTZ (45 mg/kg) were used to develop kindling seizure every 48 h for 21 days. Treated mice were administered orally fenofibrate at doses of 30 and 50 mg/kg/day during the study. Plasma corticosterone and brain levels of brain-derived neurotrophic factor (BDNF), malondialdehyde (MDA) and mRNA transcription of p53, as well as blood-brain barrier (BBB) permeability, were determined at termination of the study. Fenofibrate considerably improved seizure latency and anxiety-like behaviors in treated kindled mice. Fenofibrate at doses of 30 and 50 mg/kg significantly (P < 0.001) decreased plasma corticosterone (56.88 ± 0.80 and 54.81 ± 0.29 ng/mL, respectively) compared to PTZ group (74.96 ± 1.60 ng/mL). It also significantly (P < 0.05) decreased BDNF levels in both treatment groups (8.13 ± 0.14 and 8.74 ± 0.09 ng/mL, respectively) compared to PTZ group (9.68 ± 0.20 ng/mL). Fenofibrate particularly at higher dose significantly (P < 0.01) decreased MDA content and mRNA expression levels of p53 in treated kindled mice by 67% and 28%, respectively, compared to PTZ group. Similarly, 50 mg/kg fenofibrate significantly (P < 0.05) decreased Evans blue extravasation into brain in treated kindled mice (8.72 ± 0.96 µg/g) compared to PTZ group (15.31 ± 2.18 µg/g). Our results revealed the anticonvulsive and neuroprotective effects of fenofibrate in PTZ-induced kindling seizure in mice. Fenofibrate also improved the neurovascular functions at molecular levels in kindling seizure that might be associated with ameliorating the seizure behaviors.
Collapse
Affiliation(s)
- Nahid Sarahian
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Shamsi Darabi
- Department of Physiology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Nastaran Faghihi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
97
|
Matias Pereira AC, Sánchez-Ortíz BL, de Melo EL, da Silva Hage-Melim LI, Borges RS, Hu X, Carvalho JCT. Perillyl alcohol decreases the frequency and severity of convulsive-like behavior in the adult zebrafish model of acute seizures. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1177-1190. [PMID: 33515278 DOI: 10.1007/s00210-021-02050-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/10/2021] [Indexed: 11/28/2022]
Abstract
This research aimed to assess the effect of perillyl alcohol (PA) on convulsive behavior in vivo using adult zebrafish (Danio rerio, both sexes). The seizures were induced with pentylenetetrazole (PTZ) intraperitoneally at 170 mg/kg, and diazepam (DZP) was used as the control anticonvulsant (2 mg/kg, oral); PA was tested at 10, 50, and 100 mg/kg orally. The groups had ten animals per group (total n = 60), observed for 10 minutes after seizure induction. We manually appraised typical seizure phenotypes for quantification and used an animal tracking software (Toxtrac) to assess the motor parameters. Next, we sought to find a mechanism of action for PA anticonvulsant activity in silico using a structure-based activity prediction server and molecular docking. The results show that PTZ induced seizure-like behavior in all untreated animals with hyperlocomotion episodes, seizure itself, posture loss, and immobility. DZP inhibited the seizures in all animals of the positive control group. PA, in turn, inhibited the occurrence of seizures in a dose-dependent manner, with frequencies of 90%, 70%, and 40% (for 10, 50, and 100 mg/kg, respectively). The PA treatments also decreased several seizure endpoints in a dose-dependent manner. Also, the difference of the group treated with highest dose of PA was statistically significant compared with the negative control group for all the endpoints assessed (p < 0.05, Kruskal-Wallis). The in silico analyses suggested that PA can affect the GABAergic system, which might be involved in its anticonvulsant activity, but other mechanisms cannot be ruled out. Overall, our results suggest an anticonvulsant potential in perillyl alcohol.
Collapse
Affiliation(s)
- Arlindo César Matias Pereira
- Pharmaceutical Sciences Post-Graduation Program, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Km 02, Macapá, Amapá, CEP 68902-280, Brazil.,Drugs Research Laboratory, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Kkm 02, Macapá, Amapá, CEP 68902-280, Brazil
| | - Brenda Lorena Sánchez-Ortíz
- Drugs Research Laboratory, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Kkm 02, Macapá, Amapá, CEP 68902-280, Brazil.,Natural Products Pharmacology Laboratory, Pharmacy Department, Chemistry Faculty, National Autonomous University of Mexico, University City, Coyoacán, 04510, Mexico City, Mexico
| | - Ester Lopes de Melo
- Drugs Research Laboratory, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Kkm 02, Macapá, Amapá, CEP 68902-280, Brazil
| | - Lorane Izabel da Silva Hage-Melim
- Pharmaceutical Sciences Post-Graduation Program, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Km 02, Macapá, Amapá, CEP 68902-280, Brazil.,Laboratory of Medicinal and Pharmaceutical Chemistry (PharMedChem), Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitshek Highway, Km 02, Macapá, Amapá, CEP 68902-280, Brazil
| | - Raphaelle Sousa Borges
- Pharmaceutical Sciences Post-Graduation Program, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Km 02, Macapá, Amapá, CEP 68902-280, Brazil.,Drugs Research Laboratory, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Kkm 02, Macapá, Amapá, CEP 68902-280, Brazil
| | - Xuebo Hu
- Laboratory of Drug Discovery and Molecular Engineering, College of Plant Science and Technology, Huazhong, Agricultural University, 1 Shizishan, Nanhu, Wuhan, 430070, China
| | - José Carlos Tavares Carvalho
- Pharmaceutical Sciences Post-Graduation Program, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Km 02, Macapá, Amapá, CEP 68902-280, Brazil. .,Drugs Research Laboratory, Biological and Health Sciences Department, State University of Amapá, Juscelino Kubitisheck Highway, Kkm 02, Macapá, Amapá, CEP 68902-280, Brazil.
| |
Collapse
|
98
|
Chen S, Zou Q, Guo Q, Chen Y, Kuang X, Zhang Y, Liu Y, Wu W, Li G, Tu L, Tong J, Li S, Ma L, Li Q. SPARC Knockdown Reduces Glutamate-Induced HT22 Hippocampal Nerve Cell Damage by Regulating Autophagy. Front Neurosci 2021; 14:581441. [PMID: 33584170 PMCID: PMC7874057 DOI: 10.3389/fnins.2020.581441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein involved in the extracellular matrix and interactions between cells during neural development of the central nervous system (CNS). Oxidative glutamate toxicity is involved in CNS diseases, including epilepsy, Alzheimer’s disease, and ischemic stroke. However, the molecular mechanism of nerve injury is not fully understood in CNS diseases. Herein, the glutamate-induced nerve damage model was used to explore the molecular mechanisms affecting nerve damage. The levels of SPARC and autophagy were increased in glutamate-induced HT22 hippocampal nerve injury. In summary, the current study confirmed that SPARC regulates autophagy in HT22 hippocampal nerve cells, and its knockdown reduces the glutamate-induced HT22 hippocampal nerve injury by inhibiting autophagy. These findings suggested that SPARC plays a crucial role in nerve injury of CNS diseases.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qin Zou
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qiang Guo
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yongmin Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Xi Kuang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Hainan Health Vocational College, Haikou, China
| | - Yukang Zhang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Wengang Wu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Ge Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Linzhi Tu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Jingyi Tong
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Songrong Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Lin Ma
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qifu Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| |
Collapse
|
99
|
Pina LTS, Guimarães AG, Santos WBDR, Oliveira MA, Rabelo TK, Serafini MR. Monoterpenes as a perspective for the treatment of seizures: A Systematic Review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153422. [PMID: 33310306 DOI: 10.1016/j.phymed.2020.153422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 10/15/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Epilepsy affects more than 65 million people worldwide. Treatment for epileptic seizures is ineffective and has many adverse effects. For this reason, the search for new therapeutic options capable of filling these limitations is necessary. HYPOTHESIS/PURPOSE In this sense, natural products, such as monoterpenes, have been indicated as a new option to control neurological disorders such as epilepsy. STUDY DESIGN Therefore, the objective of this study was to review the monoterpenes that have anticonvulsive activity in animal models. METHODS The searches were performed in the PubMed, Web of Science and Scopus databases in September, 2020 and compiled studies using monoterpenes as an alternative to seizure. Two independent reviewers performed the study selection, data extraction and methodological quality assessment using the Syrcle tool. RESULTS 51 articles that described the anticonvulsant activity of 35 monoterpenes were selected with action on the main pharmacological target, including GABAA receptors, glutamate, calcium channels, sodium and potassium. In addition, these compounds are capable of reducing neuronal inflammation and oxidative stress caused by seizure. CONCLUSION These compounds stand out as a promising alternative for acting through different pharmacological mechanisms, which may not only reduce seizure, but also promote neuroprotective effect by reducing toxicity in brain regions. However, further studies are needed to determine the mechanism of action and safety assessment of these compounds.
Collapse
Affiliation(s)
- Lícia T S Pina
- Graduate Program in Health Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil.
| | - Adriana G Guimarães
- Graduate Program in Pharmaceutical Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil
| | - Wagner B da R Santos
- Graduate Program in Pharmaceutical Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil
| | - Marlange A Oliveira
- Graduate Program in Health Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil
| | - Thallita K Rabelo
- Graduate Program in Health Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil
| | - Mairim R Serafini
- Graduate Program in Health Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil; Graduate Program in Pharmaceutical Sciences, Universidade Federal de Sergipe, São Cristóvão, Sergipe, Brazil
| |
Collapse
|
100
|
Kwon HH, Lee JS, Park H, Shin J, Yin Y, Shin N, Shin HJ, Hwang JA, Kim DW, Kang JW. Vitamin E reduces spasms caused by prenatal stress by lowering calpain expression. Epilepsy Behav 2021; 114:107609. [PMID: 33257295 DOI: 10.1016/j.yebeh.2020.107609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prenatal stress increases the susceptibility of infants to seizures and is known to be associated with oxidative stress. Recent studies suggest that vitamin E has beneficial effects in various neurological diseases due to its antioxidant properties. In this study, we investigated the relationship between prenatal stress and vitamin E treatment on N-methyl-D-aspartate (NMDA)-induced spasms. METHODS We used pregnant female Sprague Dawley rats and induced prenatal stress with an injection of betamethasone on G15. They were then treated orally with 200 mg/kg vitamin E or saline twice a day from G15-G21. On postnatal day 15, NMDA was administered to trigger spasms in offspring. The total number of spasms and latency to the first spasm were recorded. We also measured oxidative stress in the medial cortex using western blot, and calpain activity, thiobarbituric acid reactive substances (TBARS), glutathione (GSH)/GSH/glutathione disulfide (GSSG), superoxide dismutase (SOD) activity, catalase activity, and nitric oxide (NO) assays. RESULTS We observed that rats treated with vitamin E while exposed to prenatal stress demonstrated reduced total number and frequency of spasms. Expression of glutamate decarboxylase 67 (GAD67) and K+/Cl- co-transporter (KCC2) were reduced after prenatal stress; this recovered in the vitamin E treated group. Further, expression of calpain 2 was decreased and various markers of oxidative stress (malondialdehyde (MDA), GSH/GSSG, SOD, catalase, and NO) were reduced in the vitamin E treated group. CONCLUSIONS Our results provide evidence that vitamin E lowers oxidative stress and decreases seizure susceptibility in rat offspring exposed to prenatal stress. Given the well-known safety profile of vitamin E, these results indicate its potential as a strategy for preventing seizures.
Collapse
Affiliation(s)
- Hyeok Hee Kwon
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Pediatrics, Chungnam National University Hospital, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Oriental Medical College of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon 301-724, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yuhua Yin
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Ah Hwang
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Anatomy, School of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Joon Won Kang
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea; Department of Pediatrics, Chungnam National University Hospital, School of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|