1
|
Marin-Castañeda LA, Pacheco Aispuro G, Gonzalez-Garibay G, Martínez Zamora CA, Romo-Parra H, Rubio-Osornio M, Rubio C. Interplay of epilepsy and long-term potentiation: implications for memory. Front Neurosci 2025; 18:1451740. [PMID: 39867454 PMCID: PMC11760605 DOI: 10.3389/fnins.2024.1451740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
The interplay between long-term potentiation (LTP) and epilepsy represents a crucial facet in understanding synaptic plasticity and memory within neuroscience. LTP, a phenomenon characterized by a sustained increase in synaptic strength, is pivotal in learning and memory processes, particularly in the hippocampus. This review delves into the intricate relationship between LTP and epilepsy, exploring how alterations in synaptic plasticity mechanisms akin to those seen in LTP contribute to the hyperexcitable state of epilepsy. This state is conceptualized as a dysregulation between LTP and LTD (Long-term depression), leading to pathologically enhanced synaptic efficacy. Additionally, the role of neuroinflammation in both LTP and epilepsy is examined, highlighting how inflammatory mediators can influence synaptic plasticity. The dual role of neuroinflammatory pathways, enhancing or inhibiting LTP, is a focal area of ongoing research. The significance of various signaling pathways, including the MAPK, mTOR, and WNT/β-catenin pathways, in the modulation of synaptic plasticity and their relevance in both LTP and epilepsy. These pathways are instrumental in memory formation, consolidation, and epileptogenesis, illustrating a complex interaction between cellular mechanisms in the nervous system. Lastly, the role of calcium signaling in the relationship between LTP and epilepsy is scrutinized. Aberrant calcium signaling in epilepsy leads to an enhanced, yet pathologically altered, LTP. This dysregulation disrupts normal neural pathways, potentially leading to cognitive dysfunction, particularly in memory encoding and retrieval. The review emphasizes the need for targeted interventions in epilepsy that address cognitive functions alongside seizure control.
Collapse
Affiliation(s)
- Luis A. Marin-Castañeda
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | | | - Guillermo Gonzalez-Garibay
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- Anahuac University, Mexico City, Mexico
| | - Carlos Alejandro Martínez Zamora
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- School of Medicine, Saint Luke, Mexico City, Mexico
| | - Hector Romo-Parra
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
- Universidad Iberoamericana, Mexico City, Mexico
| | - Moisés Rubio-Osornio
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| | - Carmen Rubio
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Mexico City, Mexico
| |
Collapse
|
2
|
Liu X, Wang FY, Chi S, Liu T, Yang HL, Zhong RJ, Li XY, Gao J. Mitochondria-targeting peptide SS-31 attenuates ferroptosis via inhibition of the p38 MAPK signaling pathway in the hippocampus of epileptic rats. Brain Res 2024; 1836:148882. [PMID: 38521160 DOI: 10.1016/j.brainres.2024.148882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Ferroptosis is a newly identified form of non-apoptotic regulated cell death (RCD) andplaysanimportantrole in epileptogenesis. The p38 mitogen-activated protein kinase (p38 MAPK) pathway has been confirmed to be involved in ferroptosis. The mitochondria-targeting antioxidant Elamipretide (SS-31) can reduce the generation of lipid peroxidation and the buildup of reactive oxygen species (ROS). Collectively, our present study was to decipher whether SS-31 inhibits ferroptosis via the p38 MAPK signaling pathway in the rat epilepsy model induced by pilocarpine (PILO).Adult male Wistar rats were randomly divided into four groups: control group (CON group), epilepsy group (EP group), SS-31 treatment group (SS group), and p38 MAPK inhibitor (SB203580) treatment group (SB group). Our results demonstrated that the rat hippocampal neurons after epilepsy were followed by accumulated iron and malondialdehyde (MDA) content, upregulated phosphorylated p38 MAPK protein (P-p38) and nuclear factor erythroid 2-related factor 2 (Nrf2) levels, reduced glutathione peroxidase 4 (Gpx4) content, and depleted glutathione (GSH) activity. Morphologically, mitochondrial ultrastructural damage under electron microscopy was manifested by a partial increase in outer membrane density, disappearance of mitochondrial cristae, and mitochondrial shrinkage. SS-31 and SB203580 treatment blocked the initiation and progression of ferroptosis in the hippocampus of epileptic rats via reducing the severity of epileptic seizures, reversing the expression of Gpx4, P-p38 , decreasing the levels of iron and MDA, as well as increasing the activity of GSH and Nrf2. To summarize, our findings proved that ferroptosis was coupled with the pathology of epilepsy, and SS-31 can inhibit PILO-induced seizures by preventing ferroptosis, which may be connected to the inhibition of p38 MAPK phosphorylation, highlighting the potential therapeutic value for targeting ferroptosis process in individuals with seizure-related diseases.
Collapse
Affiliation(s)
- Xue Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Fei-Yu Wang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Song Chi
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hai-Lin Yang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Ru-Jie Zhong
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiao-Yu Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
3
|
Dashtban-Moghadam E, Khodaverdian S, Dabirmanesh B, Mirnajafi-Zadeh J, Shojaei A, Mirzaie M, Choopanian P, Atabakhshi-Kashi M, Fatholahi Y, Khajeh K. Hippocampal tandem mass tag (TMT) proteomics analysis during kindling epileptogenesis in rat. Brain Res 2024; 1822:148620. [PMID: 37848119 DOI: 10.1016/j.brainres.2023.148620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Epilepsy is a neurological disorder that remains difficult to treat due to the lack of a clear molecular mechanism and incomplete understanding of involved proteins. To identify potential therapeutic targets, it is important to gain insight into changes in protein expression patterns related to epileptogenesis. One promising approach is to analyze proteomic data, which can provide valuable information about these changes. In this study, to evaluate the changes in gene expression during epileptogenesis, LC-MC2 analysis was carried out on hippocampus during stages of electrical kindling in rat models. Subsequently, progressive changes in the expression of proteins were detected as a result of epileptogenesis development. In line with behavioral kindled seizure stages and according to the proteomics data, we described epileptogenesis phases by comparing Stage3 versus Control (S3/C0), Stage5 versus Stage3 (S5/S3), and Stage5 versus Control group (S5/C0). Gene ontology analysis on differentially expressed proteins (DEPs) showed significant changes of proteins involved in immune responses like Csf1R, Aif1 and Stat1 during S3/C0, regulation of synaptic plasticity like Bdnf, Rac1, CaMK, Cdc42 and P38 during S5/S3, and nervous system development throughout S5/C0 like Bdnd, Kcc2 and Slc1a3.There were also proteins like Cox2, which were altered commonly among all three phases. The pathway enrichment analysis of DEPs was also done to discover molecular connections between phases and we have found that the targets like Csf1R, Bdnf and Cox2 were analyzed throughout all three phases were highly involved in the PPI network analysis as hub nodes. Additionally, these same targets underwent changes which were confirmed through Western blotting. Our results have identified proteomic patterns that could shed light on the molecular mechanisms underlying epileptogenesis which may allow for novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elahe Dashtban-Moghadam
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Shima Khodaverdian
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Medical Physiology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Amir Shojaei
- Department of Medical Physiology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Mirzaie
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland; Department of Applied Mathematics, Faculty of Mathematical Science, Tarbiat Modares University, Tehran, Iran
| | - Peyman Choopanian
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Mona Atabakhshi-Kashi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Nanoscience and Technology, Beijing 100190, China
| | - Yaghoub Fatholahi
- Department of Medical Physiology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Shan Y, Chen Y, Gu H, Wang Y, Sun Y. Regulatory Basis of Adipokines Leptin and Adiponectin in Epilepsy: from Signaling Pathways to Glucose Metabolism. Neurochem Res 2023; 48:2017-2028. [PMID: 36797447 PMCID: PMC10181973 DOI: 10.1007/s11064-023-03891-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
Epilepsy is a common and severe neurological disorder in which impaired glucose metabolism leads to changes in neuronal excitability that slow or promote the development of epilepsy. Leptin and adiponectin are important mediators regulating glucose metabolism in the peripheral and central nervous systems. Many studies have reported a strong association between epilepsy and these two adipokines involved in multiple signaling cascades and glucose metabolism. Due to the complex regulatory mechanisms between them and various signal activation networks, their role in epilepsy involves many aspects, including the release of inflammatory mediators, oxidative damage, and neuronal apoptosis. This paper aims to summarize the signaling pathways involved in leptin and adiponectin and the regulation of glucose metabolism from the perspective of the pathogenesis of epilepsy. In particular, we discuss the dual effects of leptin in epilepsy and the relationship between antiepileptic drugs and changes in the levels of these two adipokines. Clinical practitioners may need to consider these factors in evaluating clinical drugs. Through this review, we can better understand the specific involvement of leptin and adiponectin in the pathogenesis of epilepsy, provide ideas for further exploration, and bring about practical significance for the treatment of epilepsy, especially for the development of personalized treatment according to individual metabolic characteristics.
Collapse
Affiliation(s)
- Yisi Shan
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.,Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yeting Chen
- Department of Acupuncture, Zhangjiagang Second People's Hospital, Zhangjiagang, 215600, China
| | - Haiping Gu
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yadong Wang
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yaming Sun
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.
| |
Collapse
|
5
|
Exploring the mechanism of Cassiae semen in regulating lipid metabolism through network pharmacology and experimental validation. Biosci Rep 2023; 43:232453. [PMID: 36645186 PMCID: PMC9905789 DOI: 10.1042/bsr20221375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Multiple studies have assessed the role of Cassiae semen (CS) in regulating lipid metabolism. However, the mechanism of action of CS on non-alcoholic fatty liver disease (NAFLD) has seen rare scrutiny. OBJECTIVE The objective of this study was to explore the regulatory mechanism of CS on lipid metabolism in NAFLD. METHODS Components of CS ethanol extract (CSEE) were analyzed and identified using UPLC-Q-Orbirap HRMS. The candidate compounds of CS and its relative targets were extracted from the Traditional Chinese Medicine Systems Pharmacology, Swiss-Target-Prediction, and TargetNet web server. The Therapeutic Target Database, Genecards, Online Mendelian Inheritance in Man, and DisGeNET were searched for NAFLD targets. Binding affinity between potential core components and key targets was established employing molecular docking simulations. After that, free fatty acid (FFA)-induced HepG2 cells were used to further validate part of the network pharmacology results. RESULTS Six genes, including Caspase 3 (CASP3), phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α (PIK3CA), epidermal growth factor receptor (EGFR), and amyloid β (A4) precursor protein (APP) were identified as key targets. The mitogen-activated protein kinase (MAPK) signaling pathway was found to associate closely with CS's effect on NAFLD. Per molecular docking findings, toralactone and quinizarin formed the most stable combinations with hub genes. About 0.1 (vs. FFA, P<0.01) and 0.2 (vs. FFA, P<0.05) mg/ml CSEE decreased lipid accumulation in vitro by reversing the up-regulation of CASP3, EGFR, and APP and the down-regulation of PIK3CA. CONCLUSION CSEE can significantly reduce intracellular lipid accumulation by modulating the MAPK signaling pathway to decrease CASP3 and EGFR expression.
Collapse
|
6
|
Bazhanova E, Kozlov A. Mechanisms of apoptosis in drug-resistant epilepsy. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:43-50. [DOI: 10.17116/jnevro202212205143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
7
|
Chen R, Xue G, Hölscher C. The role of the TNFα-mediated astrocyte signaling pathway in epilepsy. ACTA EPILEPTOLOGICA 2021. [DOI: 10.1186/s42494-021-00059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractEpilepsy is a common disease in the central nervous system. There is growing evidence that epilepsy is associated with glial cells, including astrocytes. Tumor necrosis factor α (TNFα) is a “master regulator” of proinflammatory cytokine production and is secreted by microglia and astrocytes. TNFα secreted by microglia can activate astrocytes. Additionally, TNFα can regulate neuron activity and induce epilepsy by increasing the glutamate release, reducing the expression of γ-aminobutyric acid, inducing neuroinflammation and affecting the synaptic function in astrocytes. This review summarizes the signaling pathways and receptors of TNFα acting on astrocytes that are related to epilepsy and provides insights into the potential therapeutic strategies of epilepsy for clinical practice.
Collapse
|
8
|
Zou J, Yi S, Niu L, Zhou H, Lin Z, Wang Y, Huang X, Meng W, Guo Y, Qi L, Meng L. Neuroprotective Effect of Ultrasound Neuromodulation on Kainic Acid- Induced Epilepsy in Mice. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:3006-3016. [PMID: 33979280 DOI: 10.1109/tuffc.2021.3079628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Preliminary evidence suggests that low-intensity pulsed ultrasound (LIPUS) has neuroprotective effects on ischemic stroke, depression, and other conditions leading to neuronal cell death (e.g., Parkinson's disease). The purpose of this study was to investigate the neuroprotective effects of LIPUS in epileptic mice. Mice were made epileptic through kainic acid (KA) administration and then stimulated with LIPUS. The neuroprotective effect of ultrasound was evaluated by observing the latency, anxiety-like behavior, and levels of proteins related to inflammation, apoptosis, or signaling pathways. The safety of LIPUS was assessed by hematoxylin and eosin (H&E) and Nissl stainings. LIPUS prolonged the latency (Sham: 6.00 ± 0.26 days; 1-kHz pulse repetition frequency (PRF): 7.00 ± 0.31 days), improved the anxiety-like behavior, and inhibited the expression of inflammatory factors and apoptosis-related proteins. In addition, H&E and Nissl staining results confirmed that LIPUS did not damage the brain. These findings suggest that LIPUS has neuroprotective effects in mice with KA-induced epilepsy. LIPUS may offer a new therapeutic approach to epilepsy.
Collapse
|
9
|
Cao Q, Yang F, Wang H. CB2R induces a protective response against epileptic seizures through ERK and p38 signaling pathways. Int J Neurosci 2021; 131:735-744. [PMID: 32715907 DOI: 10.1080/00207454.2020.1796661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 05/09/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Epilepsy is a pivotal neurological disorder characterized by the synchronous discharging of neurons to induce momentary brain dysfunction. Temporal lobe epilepsy is the most common type of epilepsy, with seizures originating from the mesial temporal lobe. The hippocampus forms part of the mesial temporal lobe and plays a significant role in epileptogenesis; it also has a vital influence on the mental development of children. In this study, we aimed to explore the effects of CB2 receptor (CB2R) activation on ERK and p38 signaling in nerve cells of a rat epilepsy model. MATERIALS AND METHODS We treated Sprague-Dawley rats with pilocarpine to induce an epilepsy model and treated such animals with a CB2R agonist (JWH133) alone or with a CB2R antagonist (AM630). Nissl's stain showed the neuron conditon in different groups. Western blot analyzed the level of p-ERK and p-p38. RESULTS JWH133 can increase the latent period of first seizure attack and decrease the Grades IV-V magnitude ratio after the termination of SE. Nissl's stain showed JWH133 protected neurons in the hippocampus while AM630 inhibited the functioning of CB2R in neurons. Western blot analysis showed that JWH133 decreased levels of p-ERK and p-p38, which is found at increased levels in the hippocampus of our epilepsy model. In contrast, AM630 inhibited the protective function of JWH133 and also enhanced levels of p-ERK and p-p38. CONCLUSIONS CB2R activation can induce neurons proliferation and survival through activation of ERK and p38 signaling pathways.
Collapse
Affiliation(s)
- Qingjun Cao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fenghua Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Ahmed MM, Carrel AJ, Cruz Del Angel Y, Carlsen J, Thomas AX, González MI, Gardiner KJ, Brooks-Kayal A. Altered Protein Profiles During Epileptogenesis in the Pilocarpine Mouse Model of Temporal Lobe Epilepsy. Front Neurol 2021; 12:654606. [PMID: 34122302 PMCID: PMC8194494 DOI: 10.3389/fneur.2021.654606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
Epilepsy is characterized by recurrent, spontaneous seizures and is a major contributor to the global burden of neurological disease. Although epilepsy can result from a variety of brain insults, in many cases the cause is unknown and, in a significant proportion of cases, seizures cannot be controlled by available treatments. Understanding the molecular alterations that underlie or are triggered by epileptogenesis would help to identify therapeutics to prevent or control progression to epilepsy. To this end, the moderate throughput technique of Reverse Phase Protein Arrays (RPPA) was used to profile changes in protein expression in a pilocarpine mouse model of acquired epilepsy. Levels of 54 proteins, comprising phosphorylation-dependent and phosphorylation-independent components of major signaling pathways and cellular complexes, were measured in hippocampus, cortex and cerebellum of mice at six time points, spanning 15 min to 2 weeks after induction of status epilepticus. Results illustrate the time dependence of levels of the commonly studied MTOR pathway component, pS6, and show, for the first time, detailed responses during epileptogenesis of multiple components of the MTOR, MAPK, JAK/STAT and apoptosis pathways, NMDA receptors, and additional cellular complexes. Also noted are time- and brain region- specific changes in correlations among levels of functionally related proteins affecting both neurons and glia. While hippocampus and cortex are primary areas studied in pilocarpine-induced epilepsy, cerebellum also shows significant time-dependent molecular responses.
Collapse
Affiliation(s)
- Md Mahiuddin Ahmed
- Department of Neurology, University of Colorado Alzheimer's and Cognition Center, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrew J Carrel
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Yasmin Cruz Del Angel
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Jessica Carlsen
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Ajay X Thomas
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Section of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.,Section of Child Neurology, Texas Children's Hospital, Houston, TX, United States
| | - Marco I González
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katheleen J Gardiner
- Department of Pediatrics, Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Amy Brooks-Kayal
- Division of Neurology and Translational Epilepsy Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Children's Hospital Colorado, Aurora, CO, United States.,Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
11
|
Alachkar A, Ojha SK, Sadeq A, Adem A, Frank A, Stark H, Sadek B. Experimental Models for the Discovery of Novel Anticonvulsant Drugs: Focus on Pentylenetetrazole-Induced Seizures and Associated Memory Deficits. Curr Pharm Des 2020; 26:1693-1711. [PMID: 32003682 DOI: 10.2174/1381612826666200131105324] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Epilepsy is a chronic neurological disorder characterized by irregular, excessive neuronal excitability, and recurrent seizures that affect millions of patients worldwide. Currently, accessible antiepileptic drugs (AEDs) do not adequately support all epilepsy patients, with around 30% patients not responding to the existing therapies. As lifelong epilepsy treatment is essential, the search for new and more effective AEDs with an enhanced safety profile is a significant therapeutic goal. Seizures are a combination of electrical and behavioral events that can induce biochemical, molecular, and anatomic changes. Therefore, appropriate animal models are required to evaluate novel potential AEDs. Among the large number of available animal models of seizures, the acute pentylenetetrazole (PTZ)-induced myoclonic seizure model is the most widely used model assessing the anticonvulsant effect of prospective AEDs, whereas chronic PTZ-kindled seizure models represent chronic models in which the repeated administration of PTZ at subconvulsive doses leads to the intensification of seizure activity or enhanced seizure susceptibility similar to that in human epilepsy. In this review, we summarized the memory deficits accompanying acute or chronic PTZ seizure models and how these deficits were evaluated applying several behavioral animal models. Furthermore, major advantages and limitations of the PTZ seizure models in the discovery of new AEDs were highlighted. With a focus on PTZ seizures, the major biochemicals, as well as morphological alterations and the modulated brain neurotransmitter levels associated with memory deficits have been illustrated. Moreover, numerous medicinal compounds with concurrent anticonvulsant, procognitive, antioxidant effects, modulating effects on several brain neurotransmitters in rodents, and several newly developed classes of compounds applying computer-aided drug design (CADD) have been under development as potential AEDs. The article details the in-silico approach following CADD, which can be utilized for generating libraries of novel compounds for AED discovery. Additionally, in vivo studies could be useful in demonstrating efficacy, safety, and novel mode of action of AEDs for further clinical development.
Collapse
Affiliation(s)
- Alaa Alachkar
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666 Al Ain, United States.,Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United States
| | - Shreesh K Ojha
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666 Al Ain, United States.,Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United States
| | - Adel Sadeq
- College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Abdu Adem
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666 Al Ain, United States.,Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United States
| | - Annika Frank
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666 Al Ain, United States.,Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United States
| |
Collapse
|
12
|
Jia J, Jin J, Chen Q, Yuan Z, Li H, Bian J, Gui L. Eukaryotic expression, Co-IP and MS identify BMPR-1B protein-protein interaction network. Biol Res 2020; 53:24. [PMID: 32471519 PMCID: PMC7257232 DOI: 10.1186/s40659-020-00290-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/08/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND BMPR-1B is part of the transforming growth factor β super family and plays a pivotal role in ewe litter size. Functional loss of exon-8 mutations in the BMPR-1B gene (namely the FecB gene) can increase both the ewe ovulation rate and litter size. RESULTS This study constructed a eukaryotic expression system, prepared a monoclonal antibody, and characterized BMPR-1B/FecB protein-protein interactions (PPIs). Using Co-immunoprecipitation coupled to mass spectrometry (Co-IP/MS), 23 proteins were identified that specifically interact with FecB in ovary extracts of ewes. Bioinformatics analysis of selected PPIs demonstrated that FecB associated with several other BMPs, primarily via signal transduction in the ovary. FecB and its associated interaction proteins enriched the reproduction process via BMP2 and BMP4 pathways. Signal transduction was identified via Smads proteins and TGF-beta signaling pathway by analyzing the biological processes and pathways. Moreover, other target proteins (GDF5, GDF9, RhoD, and HSP 10) that interact with FecB and that are related to ovulation and litter size in ewes were identified. CONCLUSIONS In summary, this research identified a novel pathway and insight to explore the PPi network of BMPR-1B.
Collapse
Affiliation(s)
- Jianlei Jia
- grid.262246.60000 0004 1765 430XKey of Laboratory of Plateau Ecology and Agriculture, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China ,grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| | - Jipeng Jin
- grid.262246.60000 0004 1765 430XKey of Laboratory of Plateau Ecology and Agriculture, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China ,grid.411734.40000 0004 1798 5176College of Animal Science and Technology, Gansu Agricultural University, Lanzhou Gansu, 730070 China
| | - Qian Chen
- grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| | - Zan Yuan
- grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| | - Haiqin Li
- grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| | - Junhao Bian
- grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| | - Linsheng Gui
- grid.262246.60000 0004 1765 430XKey of Laboratory of Plateau Ecology and Agriculture, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China ,grid.262246.60000 0004 1765 430XCollege of Agriculture and Animal Husbandry, Qinghai University, 251#, Ningda Road, Chengbei District, Xining, Qinghai 810016 China
| |
Collapse
|
13
|
Alachkar A, Azimullah S, Lotfy M, Adeghate E, Ojha SK, Beiram R, Łażewska D, Kieć-Kononowicz K, Sadek B. Antagonism of Histamine H3 receptors Alleviates Pentylenetetrazole-Induced Kindling and Associated Memory Deficits by Mitigating Oxidative Stress, Central Neurotransmitters, and c-Fos Protein Expression in Rats. Molecules 2020; 25:molecules25071575. [PMID: 32235506 PMCID: PMC7181068 DOI: 10.3390/molecules25071575] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Histamine H3 receptors (H3Rs) are involved in several neuropsychiatric diseases including epilepsy. Therefore, the effects of H3R antagonist E177 (5 and 10 mg/kg, intraperitoneal (i.p.)) were evaluated on the course of kindling development, kindling-induced memory deficit, oxidative stress levels (glutathione (GSH), malondialdehyde (MDA), catalase (CAT), and superoxide dismutase (SOD)), various brain neurotransmitters (histamine (HA), acetylcholine (ACh), γ-aminobutyric acid (GABA)), and glutamate (GLU), acetylcholine esterase (AChE) activity, and c-Fos protein expression in pentylenetetrazole (PTZ, 40 mg/kg) kindled rats. E177 (5 and 10 mg/kg, i.p.) significantly decreased seizure score, increased step-through latency (STL) time in inhibitory avoidance paradigm, and decreased transfer latency time (TLT) in elevated plus maze (all P < 0.05). Moreover, E177 mitigated oxidative stress by significantly increasing GSH, CAT, and SOD, and decreasing the abnormal level of MDA (all P < 0.05). Furthermore, E177 attenuated elevated levels of hippocampal AChE, GLU, and c-Fos protein expression, whereas the decreased hippocampal levels of HA and ACh were modulated in PTZ-kindled animals (all P < 0.05). The findings suggest the potential of H3R antagonist E177 as adjuvant to antiepileptic drugs with an added advantage of preventing cognitive impairment, highlighting the H3Rs as a potential target for the therapeutic management of epilepsy with accompanied memory deficits.
Collapse
Affiliation(s)
- Alaa Alachkar
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Sheikh Azimullah
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Mohamed Lotfy
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 17666, UAE;
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE;
| | - Shreesh K. Ojha
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Rami Beiram
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Dorota Łażewska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688 Kraków, Poland; (D.Ł.); (K.K.-K.)
| | - Katarzyna Kieć-Kononowicz
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688 Kraków, Poland; (D.Ł.); (K.K.-K.)
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
- Correspondence: ; Tel.: +971-3-7137-512; Fax: +971-3-7672-033
| |
Collapse
|
14
|
Ye Q, Zeng C, Luo C, Wu Y. Ferrostatin-1 mitigates cognitive impairment of epileptic rats by inhibiting P38 MAPK activation. Epilepsy Behav 2020; 103:106670. [PMID: 31864943 DOI: 10.1016/j.yebeh.2019.106670] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 12/27/2022]
Abstract
Evidence indicates that ferrostain-1 (Fer-1), a specific inhibitor of ferroptosis, could ameliorate cognitive dysfunction of rats with kainic acid (KA)-induced temporal lobe epilepsy (TLE) by suppressing ferroptosis processes. Recent studies suggest that P38 mitogen-activated protein kinase (MAPK) pathway could be mediated by ferroptosis processes. The activation of P38 MAPK results in cognitive impairment by suppressing the expression of synaptic plasticity-related proteins. However, it is unclear whether Fer-1 can mitigate cognitive impairment of rats with KA-induced TLE by inhibiting P38 MAPK activation. In the present study, treatment with Fer-1 blocked the activation of P38 MAPK, which resulted in an increased expression of synaptophysin (SYP) and postsynaptic density protein 95 (PSD-95) in the hippocampus of rats with KA-induced TLE, hence, ameliorating their cognitive impairment. Also, P38 MAPK activation in the hippocampus of the rats reduced the expression of both PSD-95 and SYP proteins. Treatment of the rats with SB203580, a P38 MAPK-specific inhibitor, prevented the activation of P38 MAPK, which resulted in an increase in SYP and PSD95 protein levels in the hippocampus. These results suggest that Fer-1 could mitigate the cognitive impairment by suppressing P38 MAPK activation thus restoring the expression of synaptic proteins. Ferroptosis processes might be involved in suppressing synaptic protein expression.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China; Department of Neurology, The First Affiliated Hospital of University of South China, 69th Chuanshan Road, Hengyang, China
| | - Chunmei Zeng
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China
| | - Chun Luo
- Department of Neurology, Minzu Hospital of Guangxi Zhuang Autonomous Region, 262th East Mingxiu Road, Nanning, Guangxi, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, 6th Shuangyong Road, Nanning, China.
| |
Collapse
|
15
|
Hao J, Wang P, Pei D, Jia B, Hu Q. Rhein lysinate improves motor function in rats with spinal cord injury via inhibiting p38 MAPK pathway. Kaohsiung J Med Sci 2019; 35:765-771. [PMID: 31483087 DOI: 10.1002/kjm2.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jian Hao
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Ping Wang
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Dai‐Ping Pei
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Bin Jia
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| | - Qun‐Sheng Hu
- Department of Orthopedic SurgeryShenzhen Pingle Orthopedic Hospital Shenzhen China
| |
Collapse
|
16
|
Li M, Cui L, Feng X, Wang C, Zhang Y, Wang L, Ding Y, Zhao T. Losmapimod Protected Epileptic Rats From Hippocampal Neuron Damage Through Inhibition of the MAPK Pathway. Front Pharmacol 2019; 10:625. [PMID: 31231220 PMCID: PMC6565798 DOI: 10.3389/fphar.2019.00625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
Objective: This research aimed to validate the therapeutic effect of losmapimod and explore the underlying mechanism in its treatment of epilepsy. Methods: A rat model of epilepsy was constructed with an injection of pilocarpine. Microarray analysis was performed to screen aberrantly expressed mRNAs and activated signaling pathways between epileptic rats and normal controls. A TdT-mediated dUTP nick-end labeling (TUNEL) assay was used to identify cell apoptosis. Hippocampal cytoarchitecture was visualized with Nissl staining. The secretion of inflammatory factors as well as the marker proteins in the mitogen-activated protein kinase (MAPK) pathway were detected by Western blot. A Morris water maze navigation test evaluated the rats’ cognitive functions. Results: Activation of the MAPK signaling pathway was observed in epilepsy rats. A decrease in the MAPK phosphorylation level by application of losmapimod protected against epilepsy by reducing neuron loss. Losmapimod effectively improved memory, reduced the frequency of seizures, protected the neuron from damage, and limited the apoptosis of neurons in epilepsy rats. Conclusion: The application of losmapimod could partly reverse the development of epilepsy.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Lexiang Cui
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xuemin Feng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chao Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yinmeng Zhang
- Major in Clinical Medicine, Medical College of Nanchang University, Nanchang, China
| | - Lijie Wang
- Department of Traditional Chinese Medicine, General Hospital of FAW, Fourth Hospital of Jilin University, Changchuan, China
| | - Ying Ding
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
| | - Teng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Wang L, Song L, Chen X, Ma Y, Suo J, Shi J, Chen G. MiR-181b inhibits P38/JNK signaling pathway to attenuate autophagy and apoptosis in juvenile rats with kainic acid-induced epilepsy via targeting TLR4. CNS Neurosci Ther 2019; 25:112-122. [PMID: 29808547 PMCID: PMC6436603 DOI: 10.1111/cns.12991] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To explore the role of miR-181b in alterations of apoptosis and autophagy in the kainic acid (KA)-induced epileptic juvenile rats via modulating TLR4 and P38/JNK signaling pathway. METHODS Dual-luciferase reporter assay was performed to testify the targeting relationship between miR-181b and TLR4. After intracerebroventricular injection (i.c.v.) of KA, rats were injected with miR-181b agomir and TLR4 inhibitor (TAK-242). The TLR-4 activator lipopolysaccharide (LPS) was also administered into rats immediately after injection with miR-181b agomir. Quantitative real-time-polymerase chain reaction (qRT-PCR) was used for detections of miR-181b and TLR4 expressions, hematoxylin-eosin (HE) and Nissl staining for observation of the hippocampus morphological changes, and TUNEL staining for apoptosis analysis. Moreover, western blot was determined to detect TLR4 and P38/JNK pathway proteins, as well as autophagy- and apoptosis-related proteins. RESULTS TLR4 was identified as a direct target of miR-181b using Dual-luciferase reporter assay. KA rats injected with miR-181b agomir or TAK-242 had improved learning and memory abilities, reduced seizure severity of Racine's scale, and lessened neuron injury. Additionally, miR-181b agomir or TAK-242 could significantly inhibit P38/JNK signaling, decrease LC3II/I, Beclin-1, ATG5, ATG7, ATG12, Bax, and cleaved caspases-3, but increase p62 and Bcl-2 expression. No significances were found between KA group and KA + miR-181b + LPS group. CONCLUSION MiR-181b could inhibit P38/JNK signaling pathway via targeting TLR4, thereby exerting protective roles in attenuating autophagy and apoptosis of KA-induced epileptic juvenile rats.
Collapse
Affiliation(s)
- Li Wang
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Li‐Fang Song
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Xiao‐Yi Chen
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Yan‐Li Ma
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Jun‐Fang Suo
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Jing‐He Shi
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| | - Guo‐Hong Chen
- Department of NeurologyZhengzhou Children’s Hospital (Zhengzhou University Affiliated Children’s Hospital)ZhengzhouChina
| |
Collapse
|
18
|
Han CL, Zhao XM, Liu YP, Wang KL, Chen N, Hu W, Zhang JG, Ge M, Meng FG. Gene Expression Profiling of Two Epilepsy Models Reveals the ECM/Integrin signaling Pathway is Involved in Epiletogenesis. Neuroscience 2018; 396:187-199. [PMID: 30452975 DOI: 10.1016/j.neuroscience.2018.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
The molecular mechanisms underlying the development of epilepsy, i.e., epileptogenesis, are due to altered expression of a series of genes. Global expression profiling of temporal lobe epilepsy is confounded by a number of factors, including the variability among animal species, animal models, and tissue sampling time-points. In this study, we pooled two microarray datasets of the most used pilocarpine and kainic acid epilepsy models from the Gene Expression Omnibus database. A total of 567 known and novel genes were commonly differentially expressed across the two models. Pathway analyses demonstrated that the dysregulated genes were involved in 46 pathways. Real-time PCR and western blot analysis confirmed the activation of extracellular matrix (ECM)/integrin signaling pathways. Moreover, targeting ECM/integrin signaling inhibits astrocyte activation and promotes neuron injury in the hippocampus of epileptic mice. This study may provide a "gene/pathway database" that with further investigation can determine the mechanisms underlining epileptogenesis and the possible targets for neuron protection in the hippocampus after status epilepticus.
Collapse
Affiliation(s)
- Chun-Lei Han
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China; Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China
| | - Xue-Min Zhao
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China; Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China
| | - Yun-Peng Liu
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China; Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China
| | - Kai-Liang Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China; Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China
| | - Ning Chen
- Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Wei Hu
- Department of Neurology, University of Florida, FL 32607, USA
| | - Jian-Guo Zhang
- Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Ming Ge
- Department of Neurosurgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Fan-Gang Meng
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China; Beijing Key Laboratory of Neuromodulation, Beijing Municipal Science and Technology Commission, Beijing 100050, China.
| |
Collapse
|
19
|
Lu Y, Xu Q, Yin G, Xu W, Jiang H. Resolvin D1 inhibits the proliferation of lipopolysaccharide-treated HepG2 hepatoblastoma and PLC/PRF/5 hepatocellular carcinoma cells by targeting the MAPK pathway. Exp Ther Med 2018; 16:3603-3610. [PMID: 30233715 PMCID: PMC6143846 DOI: 10.3892/etm.2018.6651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) and hepatoblastoma are common malignant tumor types in China. The aim of the present study was to evaluate the effects of resolvin D1 (RvD1) on inflammatory factor levels and mitogen-activated protein kinase (MAPK) signaling in lipopolysaccharide (LPS)-treated liver cancer cells. First, HepG2 hepatoblastoma and PLC/PRF/5 HCC cells were cultured and treated with LPS with or without various concentrations of RvD1 (0, 0.025, 0.05, 0.1 and 0.2%). Subsequently, ELISA was performed to measure the protein levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 in the culture medium. In addition, cell proliferation of the liver cancer cells was assessed by MTT assay. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to detect the expression of TNF-α, IL-1β and IL-6 in the cultured cells. Western blotting was also performed to assess the protein expression of phosphorylated extracellular signal-related kinase (p-ERK), p-c-Jun N-terminal kinase (p-JNK) and p-p38. Compared with the control group, LPS treatment increased the protein levels of TNF-α, IL-1β and IL-6 in the culture medium, and RvD1 inhibited this increase in a concentration-dependent manner. RvD1 also reduced the LPS-induced increase in TNF-α, IL-1β, IL-6, p-ERK, p-JNK and p-p38 expression levels in liver cancer cells. LPS promoted the proliferation of liver cancer cells, while RvD1 attenuated this effect. In summary, the current findings suggest that RvD1 inhibits cell proliferation and the expression of inflammatory cytokines in LPS-treated liver cancer cells by targeting the MAPK pathway.
Collapse
Affiliation(s)
- You Lu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Qingyu Xu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Guowen Yin
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Weidong Xu
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Hao Jiang
- Department of Interventional Radiology, Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
20
|
Hooshmandi E, Motamedi F, Moosavi M, Katinger H, Zakeri Z, Zaringhalam J, Maghsoudi A, Ghasemi R, Maghsoudi N. CEPO-Fc (An EPO Derivative) Protects Hippocampus Against Aβ-induced Memory Deterioration: A Behavioral and Molecular Study in a Rat Model of Aβ Toxicity. Neuroscience 2018; 388:405-417. [DOI: 10.1016/j.neuroscience.2018.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 12/14/2022]
|
21
|
Wu DM, Zhang YT, Lu J, Zheng YL. Effects of microRNA-129 and its target gene c-Fos on proliferation and apoptosis of hippocampal neurons in rats with epilepsy via the MAPK signaling pathway. J Cell Physiol 2018; 233:6632-6643. [PMID: 29194604 DOI: 10.1002/jcp.26297] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/28/2017] [Indexed: 11/12/2022]
Abstract
This study aims to investigate the effect of microRNA-129 (miR-129) on proliferation and apoptosis of hippocampal neurons in epilepsy rats by targeting c-Fos via the MAPK signaling pathway. Thirty rats were equally classified into a model group (successfully established as chronic epilepsy models) and a normal group. Expression of miR-129, c-Fos, bax, and MAPK was detected by RT-qPCR and Western blotting. Hippocampal neurons were assigned into normal, blank, negative control (NC), miR-129 mimic, miR-129 inhibitor, siRNA-c-Fos, miR-129 inhibitor+siRNA-c-Fos groups. The targeting relationship between miR-129 and c-Fos was predicted and verified by bioinformatics websites and dual-luciferase reporter gene assay. Cell proliferation after transfection was measured by MTT assay, and cell cycle and apoptosis by flow cytometry. c-Fos is a potential target gene of miR-129. Compared with the normal group, the other six groups showed a decreased miR-129 expression; increased expression of expression of c-Fos, Bax, and MAPK; decreased proliferation; accelerated apoptosis; more cells arrested in the G1 phase; and fewer cells arrested in the S phase. Compared with the blank and NC groups, the miR-129 mimic group and the siRNA-c-Fos group showed decreased expression of c-Fos, Bax, and MAPK, increased cells proliferation, and decreased cell apoptosis, fewer cells arrested in the G1 phase and more cells arrested in the S phase. However, the miR-129 inhibitor groups showed reverse consequences. This study suggests that miR-129 could inhibit the occurrence and development of epilepsy by repressing c-Fos expression through inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P. R. China
| | - Yu-Tong Zhang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, P. R. China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P. R. China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P. R. China
| |
Collapse
|