1
|
Kelemen K, Sárosi M, Csüdör Á, Orbán-Kis K, Kelemen H, Bába L, Gáll Z, Horváth E, Katona I, Szilágyi T. Marked differences in the effects of levetiracetam and its analogue brivaracetam on microglial, astrocytic, and neuronal density in the rat model of kainic acid-induced temporal lobe epilepsy. Front Pharmacol 2025; 16:1553545. [PMID: 40115266 PMCID: PMC11922880 DOI: 10.3389/fphar.2025.1553545] [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: 12/30/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Efficient treatment of temporal lobe epilepsy (TLE) remains challenging due to limited understanding of cellular and network changes and the interference of novel antiepileptic drugs (AEDs) with tissue reorganisation. This study compared the effects of brivaracetam and levetiracetam on histological alterations in key brain regions of the epileptic circuitry, namely, the hippocampus, amygdala, piriform cortex (PC), endopiriform nucleus (EPN) and paraventricular thalamic nucleus (PVT), using the kainic acid (KA) rat model of TLE. Male Wistar rats were assigned to sham-operated (SHAM), epileptic (EPI), brivaracetam- (BRV-EPI) and levetiracetam-treated (LEV-EPI) epileptic groups. Epileptic groups received KA in the right lateral ventricle, which induced status epilepticus followed by a 3-week recovery and latent period. Rats then underwent 3 weeks of oral brivaracetam, levetiracetam or placebo treatment with continuous video monitoring for seizure analysis. Subsequently, triple fluorescent immunolabeling assessed microglial, astrocytic, and neuronal changes. The results showed a drastic increase in microglia density in the EPI and BRV-EPI groups compared to control and LEV-EPI. The BRV-EPI group displayed a significantly higher microglia density than SHAM and EPI groups in the right CA1, CA3 and left CA1 regions, bilateral amygdalae, EPN, PVT and left PC. Astrocyte density was significantly elevated in hippocampal regions of the BRV-EPI group, while neuronal density decreased. Furthermore, brivaracetam did not reduce seizure activity in this disease phase. Significance: Brivaracetam treatment increased microglial activation under epileptic conditions in vivo in all examined brain-regions participating in the epileptic circuitry, in contrast to the effects of levetiracetam, highlighting differences in AED-induced histological alterations.
Collapse
Affiliation(s)
- Krisztina Kelemen
- Department of Physiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Máté Sárosi
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
| | - Ágnes Csüdör
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
| | - Károly Orbán-Kis
- Department of Physiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
| | - Hanga Kelemen
- Translational Behavioural Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Neurosciences Division, Doctoral College, Semmelweis University, Budapest, Hungary
| | - László Bába
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Târgu Mures, Romania
| | - Zsolt Gáll
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Târgu Mures, Romania
| | - Eszter Horváth
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - István Katona
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
| | - Tibor Szilágyi
- Department of Physiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureș, Romania
| |
Collapse
|
2
|
Li L, Liu Z. SRF Facilitates Transcriptional Inhibition of Gem Expression by m6A Methyltransferase METTL3 to Suppress Neuronal Damage in Epilepsy. Mol Neurobiol 2025; 62:2903-2925. [PMID: 39190265 DOI: 10.1007/s12035-024-04396-x] [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: 11/01/2023] [Accepted: 07/21/2024] [Indexed: 08/28/2024]
Abstract
A bioinformatics analysis was conducted to screen for relevant expression datasets of the transcription factor SRF knockout mice. The aim was to investigate the relationship between SRF and m6A-related genes, predict how SRF regulates the m6A modification of GEM genes mediated by METTL3, and explore potential molecular mechanisms associated with neurotrauma. Disease gene databases such as GeneCards, DisGeNET, and Phenolyzer, and transcription factor databases TFDB and TRRUST, were used to obtain epilepsy-related genes and transcription factors. The intersection was then selected. Expression data of SRF knockout epilepsy mice were obtained from the GEO database and used to filter differentially expressed genes. Important module genes related to the disease were selected through WGCNA co-expression analysis. The intersection between these genes and the differentially expressed genes was performed, followed by PPI network analysis and GO/KEGG enrichment analysis. Furthermore, the core genes were selected using the cytoHubba plugin of the Cytoscape software. Differential expression analysis was performed on m6A-related factors in the GEO dataset, and the relationship between SRF and m6A-related factors and core genes was analyzed. The m6A binding sites of SRF with the METTL3 promoter and target gene Gem were predicted using the AnimalTFDB and SRAMP websites, respectively. We found that the transcription factor SRF may be a key gene in epilepsy during neuronal development. Further WGCNA analysis showed that 129 module genes were associated with SRF knockout epilepsy, and these differentially expressed genes were mainly enriched in the neuroactive ligand-receptor interaction pathway. The final results indicate that knocking out SRF may inhibit the transcription of METTL3, thereby inhibiting the m6A modification of Gem and leading to upregulation of Gem expression, thereby playing an important role in neuronal damage. Knocking out the SRF gene may inhibit the transcription of m6A methyltransferase METTL3, thereby inhibiting the m6A modification of GEM genes mediated by METTL3, promoting GEM gene expression, and leading to the occurrence of epilepsy-related neuron injury. Further investigation revealed that SRF overexpression can potentially enhance the transcription of METTL3, thus promoting m6A modification of GEM, resulting in downregulation of GEM expression. This process regulates oxidative stress in epileptic mouse neurons, suppresses inflammatory responses, and mitigates associated damage. Additionally, an in vitro neuronal epileptic model was established, and experimental techniques such as qRT-PCR and WB were employed to assess the expression of SRF, METTL3, and GEM in hippocampal tissues and neurons. The experimental results were consistent with our predictions, demonstrating that overexpression of SRF can inhibit the development of epilepsy-related neuronal damage. This study reveals that knockout of the SRF gene may suppress the transcription of m6A methyltransferase METTL3, thereby inhibiting m6A modification of the GEM gene mediated by METTL3 and subsequently promoting the expression of the GEM gene, leading to the occurrence of epilepsy-related neuronal damage.
Collapse
Affiliation(s)
- Lianling Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, P. R. China.
| | - Zhiguo Liu
- Department of Neurosurgery, Centtal Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, P. R. China
| |
Collapse
|
3
|
Zhao L, Qu HL, Zhang Y, Wu X, Ji QX, Zhang Z, Li D. ZL006 mitigates anxiety-like behaviors induced by closed head injury through modulation of the neural circuit from the medial prefrontal cortex to amygdala. Cereb Cortex 2024; 34:bhae237. [PMID: 38850218 DOI: 10.1093/cercor/bhae237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
Closed head injury is a prevalent form of traumatic brain injury with poorly understood effects on cortical neural circuits. Given the emotional and behavioral impairments linked to closed head injury, it is vital to uncover brain functional deficits and their driving mechanisms. In this study, we employed a robust viral tracing technique to identify the alteration of the neural pathway connecting the medial prefrontal cortex to the basolateral amygdala, and we observed the disruptions in neuronal projections between the medial prefrontal cortex and the basolateral amygdala following closed head injury. Remarkably, our results highlight that ZL006, an inhibitor targeting PSD-95/nNOS interaction, stands out for its ability to selectively reverse these aberrations. Specifically, ZL006 effectively mitigates the disruptions in neuronal projections from the medial prefrontal cortex to basolateral amygdala induced by closed head injury. Furthermore, using chemogenetic approaches, we elucidate that activating the medial prefrontal cortex projections to the basolateral amygdala circuit produces anxiolytic effects, aligning with the therapeutic potential of ZL006. Additionally, ZL006 administration effectively mitigates astrocyte activation, leading to the restoration of medial prefrontal cortex glutamatergic neuron activity. Moreover, in the context of attenuating anxiety-like behaviors through ZL006 treatment, we observe a reduction in closed head injury-induced astrocyte engulfment, which may correlate with the observed decrease in dendritic spine density of medial prefrontal cortex glutamatergic neurons.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Orthopedic Surgery, Shenyang Fifth People's Hospital, No. 188 Xingshun Street, Tiexi District, Shenyang 110122, Liaoning Province, China
| | - Hui Ling Qu
- Department of Neurology, General Hospital of Northern Theater Command, NO. 83 Wenhua Road, Shenhe District, Shenyang 110122, Liaoning Province, China
| | - Yan Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Xin Wu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Qian Xin Ji
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Zhuo Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| | - Dan Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, China
| |
Collapse
|
4
|
Dong Z, Min F, Zhang S, Zhang H, Zeng T. EGR1-Driven METTL3 Activation Curtails VIM-Mediated Neuron Injury in Epilepsy. Neurochem Res 2023; 48:3349-3362. [PMID: 37268752 DOI: 10.1007/s11064-023-03950-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/04/2023]
Abstract
Uncovering mechanisms underlying epileptogenesis aids in preventing further epilepsy progression and to lessen seizure severity and frequency. The purpose of this study is to explore the antiepileptogenic and neuroprotective mechanisms of EGR1 in neuron injuries encountered in epilepsy. Bioinformatics analysis was conducted to identify the key genes related to epilepsy. The mice were rendered epileptic using the kainic acid protocol, followed by measurement of seizure severity, high amplitude and frequency, pathological changes of hippocampal tissues and neuron apoptosis. Furthermore, an in vitro epilepsy model was constructed in the neurons isolated from newborn mice, which was then subjected to loss- and gain-of-function investigations, followed by neuron injury and apoptosis assessment. Interactions among EGR1, METTL3, and VIM were analyzed by a series of mechanistic experiments. In the mouse and cell models of epilepsy, VIM was robustly induced. However, its knockdown reduced hippocampal neuron injury and apoptosis. Meanwhile, VIM knockdown decreased inflammatory response and neuron apoptosis in vivo. Mechanistic investigations indicated that EGR1 transcriptionally activated METTL3, which in turn downregulated VIM expression through m6A modification. EGR1 activated METTL3 and reduced VIM expression, thereby impairing hippocampal neuron injury and apoptosis, preventing epilepsy progression. Taken together, this study demonstrates that EGR1 alleviates neuron injuries in epilepsy by inducing METTL3-mediated inhibition of VIM, which provides clues for the development of novel antiepileptic treatments.
Collapse
Affiliation(s)
- Zhaofei Dong
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, People's Republic of China
| | - Fuli Min
- Department of Neurology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, Guangdong Province, People's Republic of China
| | - Sai Zhang
- Department of Neurology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, Guangdong Province, People's Republic of China
| | - Huili Zhang
- Department of Neurology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, Guangdong Province, People's Republic of China
| | - Tao Zeng
- Department of Neurology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, Guangdong Province, People's Republic of China.
| |
Collapse
|
5
|
Wang H, Feng Y, Sun J, Zhang W, Han Z, Yu S, Gu Y, Cheng X, Lin Z, Na M. Methyl-CpG-Binding Domain Protein 3 Promotes Seizures by Recruiting Methyltransferase DNMT1 to Enhance TREM2 Methylation. Neurochem Res 2021; 46:2451-2462. [PMID: 34173118 DOI: 10.1007/s11064-021-03371-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 10/21/2022]
Abstract
Epilepsy represents a hazardous neurological disorder, underpinned by a pathophysiological process that is yet to be fully understood. Here, we aimed to elucidate the effect of methyl-CpG-binding domain protein 3 (MBD3) on hippocampal neuronal damage in epileptic mice by targeting the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway. The expression of MBD3 was determined by Western blot in a hippocampal neuronal culture (HNC) epileptic model established using the low Mg2+ECF culture method. The interaction between MBD3 and DNA methyltransferase 1 (DNMT1) was determined via co-immunoprecipitation and mass spectrometry analysis. Bisulfite modification and sequencing was performed to evaluate the degree of methylation of triggering receptor expressed on myeloid cells 2 (TREM2). The viability and apoptosis of hippocampal neurons were detected by CCK-8 and TUNEL assays, respectively. Finally, the effect of MBD3 was verified in vivo. MBD3 was highly expressed in the HNC model of epilepsy, with its interaction with DNMT1 found to promote the hypermethylation of TREM2 at site cg25748868. Additionally, decreased TREM2 and inhibited PI3K/Akt pathway was observed in the HNC epileptic model. Simultaneous inhibition of MBD3 and DNMT1 decreased the methylation level at cg25748868, up-regulated TREM2 expression, and activated the PI3K/Akt pathway, thereby arresting neuronal damage. Inhibition of MBD3 reduced the level of epileptic seizures, down-regulated cg25748868 methylation, activated TREM2-mediated signaling pathways, and alleviated hippocampal neuronal damage in the acute seizure mouse models. The present study unveiled that MBD3 and DNMT1 synergistically enhanced hypermethylation of cg25748868 in TREM2, and promoted the onset of epilepsy via inhibition of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Haiyang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Yumeng Feng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Jiaying Sun
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Wang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Zhibin Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Shengkun Yu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Yifei Gu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Xingbo Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Zhiguo Lin
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Meng Na
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
6
|
Pan W, Song X, Hu Q, Zhang Y. miR-485 inhibits histone deacetylase HDAC5, HIF1α and PFKFB3 expression to alleviate epilepsy in cellular and rodent models. Aging (Albany NY) 2021; 13:14416-14432. [PMID: 34021541 PMCID: PMC8202868 DOI: 10.18632/aging.203058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/05/2020] [Indexed: 01/15/2023]
Abstract
We investigated the role of microRNA (miR)-485 and its downstream signaling molecules on mediating epilepsy in cellular and rat models. We established a cellular epilepsy model by exposing hippocampal neurons to magnesium and a rat model by treating ICR mice with lithium chloride (127 mg/kg) and pilocarpine (30 mg/kg). We confirmed that miR-485 could bind and inhibit histone deacetylase 5 (HDAC5) and then measured expression of miR-485 and in mice and cells. Cells were transfected with overexpression or knockdown of miR-485, HDAC5, hypoxia-inducible factor-1alpha (HIF1α), or 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 enzyme (PFKFB3) to verify their roles in apoptosis, oxidative stress, and inflammation in epileptic hippocampal neurons. Binding relationship between miR-485, HDAC5, HIF1α, and PFKFB3 was verified. Oxidative stress and inflammation marker levels in epilepsy model mice were assessed. miR-485 was downregulated and HDAC5 was upregulated in cell and animal model of epilepsy. Seizure, neuronal apoptosis, oxidative stress (increased SOD and GSH-Px expression and decreased MDA and 8-OHdG expression) and inflammation (reduced IL-1β, TNF-α, and IL-6 expression) were reduced by miR-485 in epileptic cells. HIF1α and PFKFB3 expression was reduced by HDAC5 knockdown in cells, which was recapitulated in vivo. Thus, miR-485 alleviates neuronal damage and epilepsy by inhibiting HDAC5, HIF1α, and PFKFB3.
Collapse
Affiliation(s)
- Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Xingyu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qibo Hu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yunfeng Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
7
|
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
|
8
|
Lanciego JL, Wouterlood FG. Neuroanatomical tract-tracing techniques that did go viral. Brain Struct Funct 2020; 225:1193-1224. [PMID: 32062721 PMCID: PMC7271020 DOI: 10.1007/s00429-020-02041-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/31/2020] [Indexed: 12/29/2022]
Abstract
Neuroanatomical tracing methods remain fundamental for elucidating the complexity of brain circuits. During the past decades, the technical arsenal at our disposal has been greatly enriched, with a steady supply of fresh arrivals. This paper provides a landscape view of classical and modern tools for tract-tracing purposes. Focus is placed on methods that have gone viral, i.e., became most widespread used and fully reliable. To keep an historical perspective, we start by reviewing one-dimensional, standalone transport-tracing tools; these including today's two most favorite anterograde neuroanatomical tracers such as Phaseolus vulgaris-leucoagglutinin and biotinylated dextran amine. Next, emphasis is placed on several classical tools widely used for retrograde neuroanatomical tracing purposes, where Fluoro-Gold in our opinion represents the best example. Furthermore, it is worth noting that multi-dimensional paradigms can be designed by combining different tracers or by applying a given tracer together with detecting one or more neurochemical substances, as illustrated here with several examples. Finally, it is without any doubt that we are currently witnessing the unstoppable and spectacular rise of modern molecular-genetic techniques based on the use of modified viruses as delivery vehicles for genetic material, therefore, pushing the tract-tracing field forward into a new era. In summary, here, we aim to provide neuroscientists with the advice and background required when facing a choice on which neuroanatomical tracer-or combination thereof-might be best suited for addressing a given experimental design.
Collapse
Affiliation(s)
- Jose L Lanciego
- Neurosciences Department, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Avenue 55, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Floris G Wouterlood
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers, Location VUmc, Neuroscience Campus Amsterdam, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Up-Regulation of Trem2 Inhibits Hippocampal Neuronal Apoptosis and Alleviates Oxidative Stress in Epilepsy via the PI3K/Akt Pathway in Mice. Neurosci Bull 2019; 35:471-485. [PMID: 30684126 DOI: 10.1007/s12264-018-0324-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/01/2018] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a chronic and severe neurological disorder that has negative effects on the autonomous activities of patients. Functionally, Trem2 (triggering receptor expressed on myeloid cells-2) is an immunoglobulin receptor that affects neurological and psychiatric genetic diseases. Based on this rationale, we aimed to assess the potential role of Trem2 integration with the PI3K/Akt pathway in epilepsy. We used microarray-based gene expression profiling to identify epilepsy-related differentially-expressed genes. In a mouse hippocampal neuron model of epilepsy, neurons were treated with low-Mg2+ extracellular fluid, and the protein and mRNA expression of Trem2 were determined. Using a gain-of-function approach with Trem2, neuronal apoptosis and its related factors were assessed by flow cytometry, RT-qPCR, and Western blot analysis. In a pilocarpine-induced epileptic mouse model, the malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) content and superoxide dismutase (SOD) and glutathione-peroxidase (GSH-Px) activity in the hippocampus were determined, and the protein expression of Trem2 was measured. In addition, the regulatory effect of Trem2 on the PI3K/Akt pathway was analyzed by inhibiting this pathway in both the cell and mouse models of epilepsy. Trem2 was found to occupy a core position and was correlated with epilepsy. Trem2 was decreased in the hippocampus of epileptic mice and epileptic hippocampal neurons. Of crucial importance, overexpression of Trem2 activated the PI3K/Akt pathway to inhibit neuronal apoptosis. Moreover, activation of the PI3K/Akt pathway through over-expression of Trem2 alleviated oxidative stress, as shown by the increased expression of SOD and GSH-Px and the decreased expression of MDA and 8-OHdG. The current study defines the potential role of Trem2 in inhibiting the development of epilepsy, indicating that Trem2 up-regulation alleviates hippocampal neuronal injury and oxidative stress, and inhibits neuronal apoptosis in epilepsy by activating the PI3K/Akt pathway.
Collapse
|
10
|
Gill RS, Mirsattari SM, Leung LS. Resting state functional network disruptions in a kainic acid model of temporal lobe epilepsy. Neuroimage Clin 2016; 13:70-81. [PMID: 27942449 PMCID: PMC5133653 DOI: 10.1016/j.nicl.2016.11.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/19/2016] [Accepted: 11/01/2016] [Indexed: 12/16/2022]
Abstract
We studied the graph topological properties of brain networks derived from resting-state functional magnetic resonance imaging in a kainic acid induced model of temporal lobe epilepsy (TLE) in rats. Functional connectivity was determined by temporal correlation of the resting-state Blood Oxygen Level Dependent (BOLD) signals between two brain regions during 1.5% and 2% isoflurane, and analyzed as networks in epileptic and control rats. Graph theoretical analysis revealed a significant increase in functional connectivity between brain areas in epileptic than control rats, and the connected brain areas could be categorized as a limbic network and a default mode network (DMN). The limbic network includes the hippocampus, amygdala, piriform cortex, nucleus accumbens, and mediodorsal thalamus, whereas DMN involves the medial prefrontal cortex, anterior and posterior cingulate cortex, auditory and temporal association cortex, and posterior parietal cortex. The TLE model manifested a higher clustering coefficient, increased global and local efficiency, and increased small-worldness as compared to controls, despite having a similar characteristic path length. These results suggest extensive disruptions in the functional brain networks, which may be the basis of altered cognitive, emotional and psychiatric symptoms in TLE.
Collapse
Affiliation(s)
- Ravnoor Singh Gill
- Graduate Program in Neuroscience, Western University, London, Ontario, Canada
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
| | - Seyed M. Mirsattari
- Graduate Program in Neuroscience, Western University, London, Ontario, Canada
- Clinical Neurological Sciences, Western University, London, Ontario, Canada
- Department of Biomedical Imaging, Western University, London, Ontario, Canada
- Department of Biomedical Physics, Western University, London, Ontario, Canada
- Department of Psychology, Western University, London, Ontario, Canada
| | - L. Stan Leung
- Graduate Program in Neuroscience, Western University, London, Ontario, Canada
- Department of Physiology & Pharmacology, Western University, London, Ontario, Canada
| |
Collapse
|