1
|
Zhang L, Zhou Q, Zhang G, Wu C, Rong W, He S, Huo X, Zhang C. A novel prefrontal cortex and hippocampus combined brain slice based on in vivo diffusion tensor imaging of healthy male rats. Neurosci Lett 2025; 851:138171. [PMID: 40010598 DOI: 10.1016/j.neulet.2025.138171] [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: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
The pathway between the prefrontal cortex (PFC) and hippocampus (HPC) has been associated with various psychiatric disorders. While hippocampal brain slices are extensively utilized, their use has traditionally been constrained in studying long connectivity between PFC and HPC due to nerve fiber rupture during the slicing process. Consequently, optimizing brain slice preparation is crucial. The experiment consisted of three phases. Initially, the structural connection of the PFC-HPC pathway was examined using diffusion tensor imaging (DTI) data from healthy male rats. Subsequently, combined PFC-HPC brain slices were created through vibratome based on imaging acquisition. Finally, the morphology and electrophysiology of the combined brain slices were analyzed. DTI findings revealed numerous nerve fibers linking the two brain regions in the rat brain. Subsequently, a successful preparation of combined PFC-HPC brain slices cut at a 7 - 8° angle relative to the middle sagittal plane was achieved using a vibratome. Hematoxylin and eosin staining results confirmed that PFC-HPC fibers remained well-preserved in the combined brain slice. Electrophysiological recordings indicated that synchronized neuronal activity occurred in the HPC upon PFC stimulation, which depended on hippocampal activity and the integrity of PFC-to-HPC connectivity. A novel procedure for the successful preparation of healthy combined HPC-PFC brain slices, maintaining a complete fiber bundle connection between PFC and HPC, is proposed. This methodology enhances the understanding of the preservation of PFC-HPC connectivity in specific angled brain slice preparations, thereby facilitating neuroscience research focused on the longrange circuitry of subregions of interest.
Collapse
Affiliation(s)
- Lijun Zhang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Qian Zhou
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Guanghao Zhang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Changzhe Wu
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Wei Rong
- Orthopaedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, No.168 Litang Road, Changping District, Beijing 102218, China
| | - Shiji He
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Xiaolin Huo
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China
| | - Cheng Zhang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, NO.6 Beiertiao, Zhongguancun, Haidian District, Beijing 100190, China; School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, No.380, Huairou District, Beijing 101408, China.
| |
Collapse
|
2
|
Yaser S, Danzer SC. A Tail Yet to be Told: The Fasciola Cinereum in Epilepsy. Epilepsy Curr 2025; 25:125-127. [PMID: 39845192 PMCID: PMC11748303 DOI: 10.1177/15357597241309175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
The Fasciola Cinereum of the Hippocampal Tail as an Interventional Target in Epilepsy Jamiolkowski RM, Nguyen QA, Farrell JS, McGinn RJ, Hartmann DA, Nirschl JJ, Sanchez MI, Buch VP, Soltesz I. Nat Med . 2024 May;30(5):1292–1299. doi: 10.1038/s41591-024-02924-9. Targeted tissue ablation involving the anterior hippocampus is the standard of care for patients with drug-resistant mesial temporal lobe epilepsy. However, a substantial proportion continues to suffer from seizures even after surgery. We identified the fasciola cinereum (FC) neurons of the posterior hippocampal tail as an important seizure node in both mice and humans with epilepsy. Genetically defined FC neurons were highly active during spontaneous seizures in epileptic mice, and closed-loop optogenetic inhibition of these neurons potently reduced seizure duration. Furthermore, we specifically targeted and found the prominent involvement of FC during seizures in a cohort of 6 patients with epilepsy. In particular, targeted lesioning of the FC in a patient reduced the seizure burden present after ablation of anterior mesial temporal structures. Thus, the FC may be a promising interventional target in epilepsy.
Collapse
Affiliation(s)
- Sarah Yaser
- Anesthesia, Cincinnati Children's Hospital Medical Center Cincinnati
| | - Steve C Danzer
- Anesthesia, Cincinnati Children's Hospital Medical Center Cincinnati
| |
Collapse
|
3
|
Shen J, Liu Y, Liao X, Lv H, Li T, Shi J. A New Mouse Model for Exploring Tranexamic Acid-Associated Seizures. Anesth Analg 2025:00000539-990000000-01179. [PMID: 39964940 DOI: 10.1213/ane.0000000000007428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
BACKGROUND Tranexamic acid (TXA) plays a central role in antifibrinolytic strategies, raising intense concerns regarding TXA-associated seizures. The aim of the study was to develop an in vivo TXA-induced seizure (TIS) model and identify its features on behavior, electroencephalograph (EEG), histology, and metabolomics. METHODS Adult healthy male C57BL/6J mice (n = 42) were randomized into 7 dosage groups to receive intracerebroventricular injection of TXA (vehicle, 1, 5, 7.5, 10, 25, 50 μg/μL, n = 6 each). The dose titration was based on behavioral observation according to the Racine scale. Further, continuous video-EEG was performed in another 8 TIS mice and 5 control mice with the stereotaxic implantation of electrodes and guide cannulas. Then hippocampus samples were obtained from an additional 10 TIS mice and 9 control mice for histological evaluation and metabolomic analysis. RESULTS Behavioral observation revealed a dose-dependent relationship between the intensity of seizures and the intraventricular gradient of the agent. A dose of 2 μL 7.5 μg/μL TXA i.c.v. was confirmed as the target scheme to establish the model with intense behavioral seizures and typical spike epileptic EEG discharges. Histological exploration illustrated necrosis and loss of neurons as well as loss of Nissl granules in the CA3 area of the hippocampus. Additionally, metabolomic analysis revealed a variety of disturbed metabolic pathways related to seizures including amino acid, energy, antioxidant stress metabolism, and nerve signal regulation. CONCLUSIONS The study provided an in vivo mouse TIS model with specific features on behavioral seizures, EEG discharges, histological findings, and metabolomic pathways.
Collapse
Affiliation(s)
- Jingjia Shen
- From the Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Liu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xudong Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Lv
- From the Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Li
- From the Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Shi
- From the Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Eberhard J, Henning L, Fülle L, Knöpper K, Böhringer J, Graelmann FJ, Hänschke L, Kenzler J, Brosseron F, Heneka MT, Domingos AI, Eyerich S, Lochner M, Weighardt H, Bedner P, Steinhäuser C, Förster I. Ablation of CCL17-positive hippocampal neurons induces inflammation-dependent epilepsy. Epilepsia 2025; 66:554-568. [PMID: 39607395 PMCID: PMC11827734 DOI: 10.1111/epi.18200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE Neuronal cell death and neuroinflammation are characteristic features of epilepsy, but it remains unclear whether neuronal cell death as such is causative for the development of epileptic seizures. To test this hypothesis, we established a novel mouse line permitting inducible ablation of pyramidal neurons by inserting simian diphtheria toxin (DT) receptor (DTR) cDNA into the Ccl17 locus. The chemokine CCL17 is expressed in pyramidal CA1 neurons in adult mice controlling microglial quiescence. METHODS Seizure activity in CCL17-DTR mice was analyzed by electroencephalographic recordings following treatment with DT for 3 consecutive days. Neuroinflammation and neuronal cell death were evaluated by (immuno)histochemistry. Pharmacological inhibition of TNFR1 signaling was achieved by treatment with XPro1595, a dominant-negative inhibitor of soluble tumor necrosis factor. RESULTS Neuronal cell death was detectable 7 days (d7) after the first DT injection in heterozygous CCL17-DTR mice. Spontaneous epileptic seizures were observed in the vast majority of mice, often with an initial peak at d6-9, followed by a period of reduced activity and a gradual increase during the 1-month observation period. Microglial reactivity was overt from d5 after DT administration not only in the CA1 region but also in the CA2/CA3 area, shortly followed by astrogliosis. Reactive microgliosis and astrogliosis persisted until d30 and, together with neuronal loss and stratum radiatum shrinkage, reflected important features of human hippocampal sclerosis. Granule cell dispersion was detectable only 3 months after DT treatment. Application of XPro1595 significantly reduced chronic seizure burden without affecting the development of hippocampal sclerosis. SIGNIFICANCE In conclusion, our data demonstrate that sterile pyramidal neuronal death is sufficient to cause epilepsy in the absence of other pathological processes. The CCL17-DTR mouse line may thus be a valuable model for further mechanistic studies on epilepsy and assessment of antiseizure medication.
Collapse
Affiliation(s)
- Judith Eberhard
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical FacultyUniversity of BonnBonnGermany
- Deutsche ForschungsgemeinschaftBonnGermany
| | - Lorenz Fülle
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
- Business Development Europe Research Services, WuXi Biologics, Leverkusen, Germany
| | - Konrad Knöpper
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
- Howard Hughes Medical Institute and Department of Microbiology and ImmunologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Jana Böhringer
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Frederike J. Graelmann
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Lea Hänschke
- Molecular Developmental Biology, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Julia Kenzler
- Institute of Cellular Neurosciences, Medical FacultyUniversity of BonnBonnGermany
- Sanofi‐Aventis DeutschlandMedical Operations General Medicines in Germany, Switzerland, Austria (GSA)BerlinGermany
| | | | - Michael T. Heneka
- German Center for Neurodegenerative DiseasesBonnGermany
- Luxembourg Center for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Ana I. Domingos
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
| | - Stefanie Eyerich
- Institute for Medical Microbiology, Immunology, and HygieneTechnical University of MunichMunichGermany
- ZAUM—Center of Allergy and EnvironmentTechnical University and Helmholtz Center MunichMunichGermany
| | - Matthias Lochner
- Institute for Medical Microbiology, Immunology, and HygieneTechnical University of MunichMunichGermany
- Institute of Medical Microbiology and Hospital EpidemiologyHannover Medical SchoolHannoverGermany
| | - Heike Weighardt
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical FacultyUniversity of BonnBonnGermany
| | | | - Irmgard Förster
- Immunology & Environment, Life and Medical Sciences InstituteUniversity of BonnBonnGermany
| |
Collapse
|
5
|
Huang X, Zhang Y, Lin Q, Huang K, Li Y, Liu P, Cao D, Li W, Li W, Li X, Gong Q, Zhou D, An D. Association of valproate use and hippocampal atrophy in idiopathic generalized epilepsy. Neuroimage Clin 2025; 45:103744. [PMID: 39914290 PMCID: PMC11847523 DOI: 10.1016/j.nicl.2025.103744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/03/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025]
Abstract
OBJECTIVE Recent studies revealed the effect of valproate (VPA) on brain structural changes in idiopathic generalized epilepsy (IGE). We aimed to investigate the volume of the entire hippocampus and subfields in patients with IGE, and explored their associations with VPA use. METHODS A total of 211 patients with IGE and 97 healthy controls (HCs) were enrolled in this study. All participants underwent T1-weighted images. Each hippocampus was segmented into seven subfields using HippUnfold. The volumes of bilateral hippocampi and each hippocampal subfield were evaluated. Spearman correlation analyses were performed to identify VPA use related abnormalities in IGE. Subgroup analyses for juvenile myoclonic epilepsy (JME), epilepsy with generalized tonic-clonic seizures alone (GTCA), and absence epilepsy (AE) were conducted. RESULTS The volumes of bilateral hippocampi were reduced in IGE compared with HCs. Subgroup analysis showed significant volume reductions in right hippocampus and its subfields in GTCA. Additionally, significant volume reductions were detected in bilateral hippocampal volumes and subfields in IGE patients currently taking VPA compared with HCs. A negative correlation was observed between the left CA2 volume and the age of onset. CONCLUSIONS Our study revealed volume reductions in bilateral hippocampi in IGE, as well as in the right hippocampus and its subfields in GTCA. Abnormalities in both subfields and the whole hippocampus were associated with VPA use. These findings suggest that VPA may have more extensive neuroanatomical effects in IGE, potentially accounting for the heterogeneity observed in neuroimaging studies.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingying Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuxing Lin
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kailing Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuming Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiwen Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyang Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenhao Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Li
- Department of Geriatric Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiuli Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Dongmei An
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Shih YT, Alipio JB, Klaft ZJ, Green N, Wong LP, Sadreyev R, Hyun JH, Dulla C, Sahay A. Pro-cognitive restoration of experience-dependent parvalbumin inhibitory neuron plasticity in neurodevelopmental disorders. RESEARCH SQUARE 2025:rs.3.rs-5624085. [PMID: 39877097 PMCID: PMC11774442 DOI: 10.21203/rs.3.rs-5624085/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The hippocampus forms memories of our experiences by registering processed sensory information in coactive populations of excitatory principal cells or ensembles1-3. Fast-spiking parvalbumin-expressing inhibitory neurons (PV INs) in the dentate gyrus (DG)-CA3/CA2 circuit contribute to memory encoding by exerting precise temporal control of excitatory principal cell activity through mossy fiber-dependent feed-forward inhibition4-13. PV INs respond to input-specific information by coordinating changes in their intrinsic excitability, input-output synaptic-connectivity, synaptic-physiology and synaptic-plasticity9,13-17, referred to here as experience-dependent PV IN plasticity, to influence hippocampal functions. PV IN impairments in early life, when neural circuitry is highly sensitive to experience-dependent refinement, are thought to result in imbalanced excitation and inhibition, impaired cognition, network hyperexcitability and seizures: hallmarks of neurodevelopmental disorders (NDDs) such as Autism Spectrum Disorder and epilepsy18-20. Discovery of transcriptional regulators of experience-dependent PV IN plasticity in the adult hippocampus may permit reversal of these developmental impairments. Here, in a screen designed to capture the PV IN intrinsic program induced by increased mossy fiber inputs, a trigger for experience-dependent PV IN plasticity, we identify the homeobox gene Meis2 21 as a regulator of experience-dependent PV IN plasticity gene (XPG) in the adult DG-CA3/CA2 circuit. We found that a significant number of upregulated XPGs also exhibit haploinsufficiency in ASDs, epilepsies, and schizophrenia. We demonstrate that virally-mediated rescue of experience-dependent Meis2 upregulation in CA3/CA2 PV INs in a NDD risk mouse model in adulthood is sufficient to restore experience-dependent PV IN plasticity, spatial and social memory, ensemble specificity, suppression of network hyperexcitability and seizures. Together, these findings suggest that experience-dependent PV IN plasticity is a convergent mechanism for NDD risk genes that can be re-instated in adulthood to reverse developmental deficits in circuitry, network excitability and cognition.
Collapse
Affiliation(s)
- Yu-Tzu Shih
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zin-Juan Klaft
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Nathaniel Green
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lai Ping Wong
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Chris Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
7
|
Duan X, Zhang C, Wu Y, Ju J, Xu Z, Li X, Liu Y, Ohdah S, Constantin OM, Pan Y, Lu Z, Wang C, Chen X, Gee CE, Nagel G, Hou ST, Gao S, Song K. Suppression of epileptic seizures by transcranial activation of K +-selective channelrhodopsin. Nat Commun 2025; 16:559. [PMID: 39789018 PMCID: PMC11718177 DOI: 10.1038/s41467-025-55818-w] [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: 06/12/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Optogenetics is a valuable tool for studying the mechanisms of neurological diseases and is now being developed for therapeutic applications. In rodents and macaques, improved channelrhodopsins have been applied to achieve transcranial optogenetic stimulation. While transcranial photoexcitation of neurons has been achieved, noninvasive optogenetic inhibition for treating hyperexcitability-induced neurological disorders has remained elusive. There is a critical need for effective inhibitory optogenetic tools that are highly light-sensitive and capable of suppressing neuronal activity in deep brain tissue. In this study, we developed a highly sensitive moderately K+-selective channelrhodopsin (HcKCR1-hs) by molecular engineering of the recently discovered Hyphochytrium catenoides kalium (potassium) channelrhodopsin 1. Transcranial activation of HcKCR1-hs significantly prolongs the time to the first seizure, increases survival, and decreases seizure activity in several status epilepticus mouse models. Our approach for transcranial optogenetic inhibition of neural hyperactivity may be adapted for cell type-specific neuromodulation in both basic and preclinical settings.
Collapse
Affiliation(s)
- Xiaodong Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Chong Zhang
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Yujie Wu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jun Ju
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhe Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xuanyi Li
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yao Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Schugofa Ohdah
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Oana M Constantin
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Yifan Pan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghua Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Christine E Gee
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Sheng-Tao Hou
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany.
| | - Kun Song
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
8
|
Colmers PLW, Antonoudiou P, Basu T, Coleman EM, He Y, Scapa G, Fuller P, Maguire J. Loss of PV Interneurons in the BLA May Contribute to Altered Network and Behavioral States in Chronically Epileptic Mice. eNeuro 2025; 12:ENEURO.0482-23.2024. [PMID: 39746805 PMCID: PMC11773627 DOI: 10.1523/eneuro.0482-23.2024] [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: 11/18/2023] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Psychiatric disorders, including anxiety and depression, are highly comorbid in people with epilepsy. However, the mechanisms mediating the shared pathophysiology are currently unknown. There is considerable evidence implicating the basolateral amygdala (BLA) in the network communication of anxiety and fear, a process demonstrated to involve parvalbumin-positive (PV) interneurons. The loss of PV interneurons has been well described in the hippocampus of chronically epileptic mice and in postmortem human tissue of patients with temporal lobe epilepsy (TLE). We hypothesize that a loss of PV interneurons in the BLA may contribute to comorbid mood disorders in epilepsy. To test this hypothesis, we employed a ventral intrahippocampal kainic acid model of TLE in mice, which exhibits profound behavioral deficits associated with chronic epilepsy. We demonstrate a loss of PV interneurons and dysfunction of the remaining PV interneurons in the BLA of chronically epileptic mice. Furthermore, we demonstrate altered principal neuron function and impaired coordination of BLA network and behavioral states in chronically epileptic mice. To determine whether the loss of PV interneurons contributes to these altered network and behavioral states, we partially ablated PV interneurons in the BLA by stereotaxically injecting AAV-Flex-DTA into the BLA of PV-Cre mice. Loss of PV interneurons in the BLA is sufficient to alter behavioral states, such as increasing avoidance behaviors and impairing fear learning. These data suggest that compromised inhibition in the BLA in chronically epileptic mice may contribute to behavioral deficits, suggesting a novel mechanism contributing to comorbid anxiety and epilepsy.
Collapse
Affiliation(s)
- Phillip L W Colmers
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Pantelis Antonoudiou
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Trina Basu
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Emanuel M Coleman
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Yingchu He
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Garrett Scapa
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Patrick Fuller
- Department of Neurological Surgery, UC Davis Health, Sacramento, California 95817
| | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
9
|
Li Z, Lu W, Yang L, Lai N, Wang Y, Chen Z. Decade of TRAP progress: Insights and future prospects for advancing functional network research in epilepsy. Prog Neurobiol 2025; 244:102707. [PMID: 39725016 DOI: 10.1016/j.pneurobio.2024.102707] [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: 08/01/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Targeted Recombination in Active Populations (TRAP) represents an effective and extensively applied technique that has earned significant utilization in neuroscience over the past decade, primarily for identifying and modulating functionally activated neuronal ensembles associated with diverse behaviors. As epilepsy is a neurological disorder characterized by pathological hyper-excitatory networks, TRAP has already been widely applied in epilepsy research. However, the deployment of TRAP in this field remains underexplored, and there is significant potential for further application and development in epilepsy-related investigations. In this review, we embark on a concise examination of the mechanisms behind several TRAP tools, introduce the current applications of TRAP in epilepsy research, and collate the key advantages as well as limitations of TRAP. Furthermore, we sketch out perspectives on potential applications of TRAP in future epilepsy research, grounded in the present landscape and challenges of the field, as well as the ways TRAP has been embraced in other neuroscience domains.
Collapse
Affiliation(s)
- Zhisheng Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wangjialu Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Yang
- key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
10
|
Baudouin SJ, Giles AR, Pearson N, Deforges S, He C, Boileau C, Partouche N, Borta A, Gautron J, Wartel M, Bočkaj I, Scavarda D, Bartolomei F, Penchet G, Aupy J, Sims J, Smith J, Mercer A, Danos O, Mulle C, Crépel V, Porter R. A novel AAV9-dual microRNA-vector targeting GRIK2 in the hippocampus as a treatment for mesial temporal lobe epilepsy. Mol Ther Methods Clin Dev 2024; 32:101342. [PMID: 39429724 PMCID: PMC11489344 DOI: 10.1016/j.omtm.2024.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024]
Abstract
Mesial temporal lobe epilepsy (mTLE) is the most prevalent type of epilepsy in adults. First and subsequent generations of anti-epileptic therapy regimens fail to decrease seizures in a large number of patients suffering from mTLE, leaving surgical ablation of part of the hippocampus as the only therapeutic option to potentially reach seizure freedom. GluK2 has recently been identified as a promising target for the treatment of mTLE using gene therapy. Here, we engineered an adeno-associated virus serotype 9 vector expressing a cluster of two synthetic microRNAs (miRNAs), expressed from the human synapsin promoter, that target GRIK2 mRNA. Intra-hippocampal delivery of this vector in a mouse model of mTLE significantly reduced GRIK2 expression and daily seizure frequency. This treatment also improved the animals' health, reduced their anxiety, and restored working memory. Focal administration of the vector to the hippocampus of cynomolgus monkeys in GLP toxicology studies led to the selective transduction of hippocampal neurons with little exposure elsewhere in the brain and no transduction outside the central nervous system. Expression of miRNAs in hippocampal neurons resulted in substantially decreased GRIK2 mRNA expression. These data suggest that the intra-hippocampal delivery of a GMP-grade AAV9 encoding a synthetic miRNAs targeting GRIK2 is a promising treatment strategy for mTLE.
Collapse
Affiliation(s)
| | | | - Nick Pearson
- uniQure (Corlieve Therapeutics AG), 4052 Basel, Switzerland
| | | | - Chenxia He
- uniQure (Corlieve Therapeutics AG), 4052 Basel, Switzerland
| | - Céline Boileau
- INSERM, INMED, Aix-Marseille University, 13009 Marseille, France
| | | | - Andreas Borta
- uniQure (Corlieve Therapeutics AG), 4052 Basel, Switzerland
| | | | - Morgane Wartel
- uniQure biopharma B.V., 1105BP Amsterdam, the Netherlands
| | - Irena Bočkaj
- uniQure biopharma B.V., 1105BP Amsterdam, the Netherlands
| | - Didier Scavarda
- APHM, INSERM, Aix-Marseille University, Timone Hospital, Pediatric Neurosurgery, 13005 Marseille, France
| | - Fabrice Bartolomei
- APHM, INSERM, Aix-Marseille University, INS, Timone Hospital, Epileptology Department, 13005 Marseille, France
| | - Guillaume Penchet
- Pellegrin Hospital, Neurosurgery Department, CHU, 33000 Bordeaux, France
| | - Jérôme Aupy
- Pellegrin Hospital, Neurosurgery Department, CHU, 33000 Bordeaux, France
| | | | | | | | | | | | - Valérie Crépel
- INSERM, INMED, Aix-Marseille University, 13009 Marseille, France
| | - Richard Porter
- uniQure (Corlieve Therapeutics AG), 4052 Basel, Switzerland
| |
Collapse
|
11
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. eLife 2024; 12:RP90893. [PMID: 39446467 PMCID: PMC11501206 DOI: 10.7554/elife.90893] [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] [Indexed: 10/26/2024] Open
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2Baxfl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild-type females or males, which is notable because loss of these two hilar cell types is implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, University of SaskatchewanSaskatoonCanada
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Neuroscience & Physiology, Psychiatry, and the Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
12
|
Zouridis IS, Balsamo G, Preston-Ferrer P, Burgalossi A. Anatomical and electrophysiological analysis of the fasciola cinerea of the mouse hippocampus. Hippocampus 2024; 34:528-539. [PMID: 39105449 DOI: 10.1002/hipo.23623] [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: 06/13/2023] [Revised: 05/20/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024]
Abstract
The hippocampus is considered essential for several forms of declarative memory, including spatial and social memory. Despite the extensive research of the classic subfields of the hippocampus, the fasciola cinerea (FC)-a medially located structure within the hippocampal formation-has remained largely unexplored. In the present study, we performed a morpho-functional characterization of principal neurons in the mouse FC. Using in vivo juxtacellular recording of single neurons, we found that FC neurons are distinct from neighboring CA1 pyramidal cells, both morphologically and electrophysiologically. Specifically, FC neurons displayed non-pyramidal morphology and granule cell-like apical dendrites. Compared to neighboring CA1 pyramidal neurons, FC neurons exhibited more regular in vivo firing patterns and a lower tendency to fire spikes at short interspike intervals. Furthermore, tracing experiments revealed that the FC receives inputs from the lateral but not the medial entorhinal cortex and CA3, and it provides a major intra-hippocampal projection to the septal CA2 and sparser inputs to the distal CA1. Overall, our results indicate that the FC is a morphologically and electrophysiologically distinct subfield of the hippocampal formation; given the established role of CA2 in social memory and seizure initiation, the unique efferent intra-hippocampal connectivity of the FC points to possible roles in social cognition and temporal lobe epilepsy.
Collapse
Affiliation(s)
- Ioannis S Zouridis
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Giuseppe Balsamo
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Patricia Preston-Ferrer
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Andrea Burgalossi
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.08.548217. [PMID: 37502909 PMCID: PMC10369878 DOI: 10.1101/2023.07.08.548217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2 Bax fl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were the more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild type females or males, which is notable because these two hilar cell types are implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, The University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
- Departments of Neuroscience & Physiology, Psychiatry, and the New York University, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
14
|
Liu X, Zhao Y, Liang X, Ding Y, Hu J, Deng N, Zhao Y, Huang P, Xie W. In Vivo Evaluation of Self-assembled nano-Saikosaponin-a for Epilepsy Treatment. Mol Biotechnol 2024; 66:2230-2240. [PMID: 37608078 DOI: 10.1007/s12033-023-00851-7] [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: 04/10/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Saikosaponin-a (SSa) exhibits antiepileptic effects. However, its poor water solubility and inability to pass through the blood-brain barrier greatly limit its clinical development and application. In this study, SSa-loaded Methoxy poly (ethylene glycol)-poly(ε-caprolactone) (MePEG-SSa-PCL) NPs were successfully prepared and characterized. Our objective was to further investigate the effect of this composite on acute seizure in mice. First, we confirmed the particle size and surface potential of the composite (51.00 ± 0.25 nm and - 33.77 ± 2.04 mV, respectively). Further, we compared the effects of various MePEG-SSa-PCL doses (low, medium, and high) with those of free SSa, valproic acid (VPA - positive control), and saline only (model group) on acute seizure using three different acute epilepsy mouse models. We observed that compared with the model group, the three MePEG-SSa-PCL treatments showed significantly lowered seizure frequency in mice belonging to the maximum electroconvulsive model group. In the pentylenetetrazol and kainic acid (KA) acute epilepsy models, MePEG-SSa-PCL increased both clonic and convulsion latency periods and shortened convulsion duration more effectively than equivalent SSa-only doses. Furthermore, hematoxylin-eosin and Nissl staining revealed considerably less neuronal damage in the hippocampal CA3 area of KA mice in the SSa, VPA, and three MePEG-SSa-PCL groups relative to mice in the model group. Hippocampal gamma-aminobutyric acid-A (GABA-A) receptor and cleaved caspase-3 expression levels in KA mice were significantly higher and lower, respectively, in the three MePEG-SSa-PCL treatment groups than in the model group. Thus, MePEG-SSa-PCL exhibited a more potent antiepileptic effect than SSa in acute mouse epilepsy models and could alleviate neuronal damage in the hippocampus following epileptic seizures, possibly via GABA-A receptor expression upregulation.
Collapse
Affiliation(s)
- Xueqi Liu
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yunyan Zhao
- Department of Critical Care Medicine, The Afflliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, 510130, China
| | - Xiaoshan Liang
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yuewen Ding
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiao Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Ning Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yiting Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Ping Huang
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wei Xie
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Lu T, Chen X, Zhang Q, Shang K, Yang X, Xiang W. Vitamin D Relieves Epilepsy Symptoms and Neuroinflammation in Juvenile Mice by Activating the mTOR Signaling Pathway via RAF1: Insights from Network Pharmacology and Molecular Docking Studies. Neurochem Res 2024; 49:2379-2392. [PMID: 38837094 DOI: 10.1007/s11064-024-04176-y] [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: 11/10/2023] [Revised: 03/15/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Epilepsy is a common neurological disorder, and the exploration of potential therapeutic drugs for its treatment is still ongoing. Vitamin D has emerged as a promising treatment due to its potential neuroprotective effects and anti-epileptic properties. This study aimed to investigate the effects of vitamin D on epilepsy and neuroinflammation in juvenile mice using network pharmacology and molecular docking, with a focus on the mammalian target of rapamycin (mTOR) signaling pathway. Experimental mouse models of epilepsy were established through intraperitoneal injection of pilocarpine, and in vitro injury models of hippocampal neurons were induced by glutamate (Glu) stimulation. The anti-epileptic effects of vitamin D were evaluated both in vivo and in vitro. Network pharmacology and molecular docking analysis were used to identify potential targets and regulatory pathways of vitamin D in epilepsy. The involvement of the mTOR signaling pathway in the regulation of mouse epilepsy by vitamin D was validated using rapamycin (RAPA). The levels of inflammatory cytokines (TNF-α, IL-1β, and IL-6) were assessed by enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. The terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) staining was used to analyze the apoptosis of hippocampal neurons. In in vivo experiments, vitamin D reduced the Racine scores of epileptic mice, prolonged the latency of epilepsy, and inhibited the production of TNF-α, IL-1β, and IL-6 in the hippocampus. Furthermore, network pharmacology analysis identified RAF1 as a potential target of vitamin D in epilepsy, which was further confirmed by molecular docking analysis. Additionally, the mTOR signaling pathway was found to be involved in the regulation of mouse epilepsy by vitamin D. In in vitro experiments, Glu stimulation upregulated the expressions of RAF1 and LC3II/LC3I, inhibited mTOR phosphorylation, and induced neuronal apoptosis. Mechanistically, vitamin D activated the mTOR signaling pathway and alleviated mouse epilepsy via RAF1, while the use of the pathway inhibitor RAPA reversed this effect. Vitamin D alleviated epilepsy symptoms and neuroinflammation in juvenile mice by activating the mTOR signaling pathway via RAF1. These findings provided new insights into the molecular mechanisms underlying the anti-epileptic effects of vitamin D and further supported its use as an adjunctive therapy for existing anti-epileptic drugs.
Collapse
Affiliation(s)
- Tiantian Lu
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Xiuling Chen
- Department of Pediatric Medicine, Affiliated Haikou Hospital of Xiangya Medical School Central South University, Haikou, 570208, China
| | - Qin Zhang
- Department of Neurosurgery, Hainan Women and Children's Medical Center, Haikou, 570312, China
| | - Kun Shang
- Institute of Deep-sea Science and Engineering, Chinese Academy of Science, Sanya, 572000, China
| | - Xiaogui Yang
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Wei Xiang
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China.
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570312, China.
- National Health Commission (NHC) Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, 570216, China.
| |
Collapse
|
16
|
Wang Y, Yang H, Li N, Wang L, Guo C, Ma W, Liu S, Peng C, Chen J, Song H, Chen H, Ma X, Yi J, Lian J, Kong W, Dong J, Tu X, Shah M, Tian X, Huang Z. A Novel Ubiquitin Ligase Adaptor PTPRN Suppresses Seizure Susceptibility through Endocytosis of Na V1.2 Sodium Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400560. [PMID: 38874331 PMCID: PMC11304301 DOI: 10.1002/advs.202400560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Indexed: 06/15/2024]
Abstract
Intrinsic plasticity, a fundamental process enabling neurons to modify their intrinsic properties, plays a crucial role in shaping neuronal input-output function and is implicated in various neurological and psychiatric disorders. Despite its importance, the underlying molecular mechanisms of intrinsic plasticity remain poorly understood. In this study, a new ubiquitin ligase adaptor, protein tyrosine phosphatase receptor type N (PTPRN), is identified as a regulator of intrinsic neuronal excitability in the context of temporal lobe epilepsy. PTPRN recruits the NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L) to NaV1.2 sodium channels, facilitating NEDD4L-mediated ubiquitination, and endocytosis of NaV1.2. Knockout of PTPRN in hippocampal granule cells leads to augmented NaV1.2-mediated sodium currents and higher intrinsic excitability, resulting in increased seizure susceptibility in transgenic mice. Conversely, adeno-associated virus-mediated delivery of PTPRN in the dentate gyrus region decreases intrinsic excitability and reduces seizure susceptibility. Moreover, the present findings indicate that PTPRN exerts a selective modulation effect on voltage-gated sodium channels. Collectively, PTPRN plays a significant role in regulating intrinsic excitability and seizure susceptibility, suggesting a potential strategy for precise modulation of NaV1.2 channels' function.
Collapse
Affiliation(s)
- Yifan Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Na Li
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weining Ma
- Department of NeurologyShengjing Hospital Affiliated to China Medical UniversityShenyang110022China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jiexin Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hedan Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingyun Yi
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingjing Lian
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Mala Shah
- UCL School of PharmacyUniversity College LondonLondonWC1N 1AXUK
| | - Xin Tian
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of NeurologyChongqing400016China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| |
Collapse
|
17
|
Suleiman Khoury Z, Sohail F, Wang J, Mendoza M, Raake M, Tahoor Silat M, Reddy Bathinapatta M, Sadeghzadegan A, Meghana P, Paul J. Neuroinflammation: A Critical Factor in Neurodegenerative Disorders. Cureus 2024; 16:e62310. [PMID: 39006715 PMCID: PMC11246070 DOI: 10.7759/cureus.62310] [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] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
This review offers a comprehensive review of the signals and the paramount role neuroinflammation plays in neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. The study explores the sophisticated interactions between microglial, astrocytic, and dendritic cells and how neuroinflammation affects long-term neuronal damage and dysfunction. There are specific pathways related to the mentioned inflammatory processes, including Janus kinases/signal transducer and activator of transcriptions, nuclear factor-κB, and mitogen-activated protein kinases pathways. Neuroinflammation is argued to be a double-edged sword, being not only a protective agent that prevents further neuron damage but also the causative factor in more cell injury development. This concept of contrasting inflammation with neuroprotection advocates for the use of therapeutic techniques that seek to modulate neuroinflammatory responses as part of the neurodegeneration treatment. The recent research findings are integrated with the established knowledge to help present a comprehensive image of neuroinflammation's impact on neurodegenerative diseases and its implications for future therapy.
Collapse
Affiliation(s)
| | - Fatima Sohail
- Department of Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, USA
| | - Jada Wang
- Department of Medicine, St. George's University, Brooklyn, USA
| | - Moises Mendoza
- Department of Health Sciences, Universidad Centroccidental Lisandro Alvarado, Barquisimeto, VEN
| | - Mohammed Raake
- Department of Medicine, Annamalai University, Chennai, IND
| | | | | | - Amirali Sadeghzadegan
- Department of General Practice, Marmara University School of Medicine, Istanbul, TUR
| | - Patel Meghana
- Department of Medicine, Ramaiah University of Applied Sciences, Bengaluru, IND
| | - Janisha Paul
- Department of Medicine, Punjab Institute of Medical Sciences, Jalandhar, IND
| |
Collapse
|
18
|
B Szabo A, Sayegh F, Gauzin S, Lejards C, Guiard B, Valton L, Verret L, Rampon C, Dahan L. No major effect of dopamine receptor 1/5 antagonist SCH-23390 on epileptic activity in the Tg2576 mouse model of amyloidosis. Eur J Neurosci 2024; 59:1558-1566. [PMID: 38308520 DOI: 10.1111/ejn.16268] [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: 08/01/2023] [Revised: 12/19/2023] [Accepted: 01/14/2024] [Indexed: 02/04/2024]
Abstract
The excitation-inhibition imbalance manifesting as epileptic activities in Alzheimer's disease is gaining more and more attention, and several potentially involved cellular and molecular pathways are currently under investigation. Based on in vitro studies, dopamine D1-type receptors in the anterior cingulate cortex and the hippocampus have been proposed to participate in this peculiar co-morbidity in mouse models of amyloidosis. Here, we tested the implication of dopaminergic transmission in vivo in the Tg2576 mouse model of Alzheimer's disease by monitoring epileptic activities via intracranial EEG before and after treatment with dopamine antagonists. Our results show that neither the D1-like dopamine receptor antagonist SCH23390 nor the D2-like dopamine receptor antagonist haloperidol reduces the frequency of epileptic activities. While requiring further investigation, our results indicate that on a systemic level, dopamine receptors are not significantly contributing to epilepsy observed in vivo in this mouse model of Alzheimer's disease.
Collapse
Affiliation(s)
- Anna B Szabo
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- Centre de recherche Cerveau et Cognition (CerCo), CNRS, UMR 5549, Toulouse Mind and Brain Institute (TMBI), University of Toulouse, University Paul Sabatier (UPS), Toulouse, France
| | - Farès Sayegh
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sèbastien Gauzin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Camille Lejards
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Bruno Guiard
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Luc Valton
- Centre de recherche Cerveau et Cognition (CerCo), CNRS, UMR 5549, Toulouse Mind and Brain Institute (TMBI), University of Toulouse, University Paul Sabatier (UPS), Toulouse, France
- Department of Neurology, Hôpital Pierre Paul Riquet - Purpan, Toulouse University Hospital, University of Toulouse, Toulouse, France
| | - Laure Verret
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
19
|
Xiao W, Li P, Kong F, Kong J, Pan A, Long L, Yan X, Xiao B, Gong J, Wan L. Unraveling the Neural Circuits: Techniques, Opportunities and Challenges in Epilepsy Research. Cell Mol Neurobiol 2024; 44:27. [PMID: 38443733 PMCID: PMC10914928 DOI: 10.1007/s10571-024-01458-5] [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: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024]
Abstract
Epilepsy, a prevalent neurological disorder characterized by high morbidity, frequent recurrence, and potential drug resistance, profoundly affects millions of people globally. Understanding the microscopic mechanisms underlying seizures is crucial for effective epilepsy treatment, and a thorough understanding of the intricate neural circuits underlying epilepsy is vital for the development of targeted therapies and the enhancement of clinical outcomes. This review begins with an exploration of the historical evolution of techniques used in studying neural circuits related to epilepsy. It then provides an extensive overview of diverse techniques employed in this domain, discussing their fundamental principles, strengths, limitations, as well as their application. Additionally, the synthesis of multiple techniques to unveil the complexity of neural circuits is summarized. Finally, this review also presents targeted drug therapies associated with epileptic neural circuits. By providing a critical assessment of methodologies used in the study of epileptic neural circuits, this review seeks to enhance the understanding of these techniques, stimulate innovative approaches for unraveling epilepsy's complexities, and ultimately facilitate improved treatment and clinical translation for epilepsy.
Collapse
Affiliation(s)
- Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Peile Li
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Fujiao Kong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jingyi Kong
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children's Hospital, Changsha, Hunan Province, China.
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China.
| |
Collapse
|
20
|
Basu S, Ro EJ, Liu Z, Kim H, Bennett A, Kang S, Suh H. The Mef2c Gene Dose-Dependently Controls Hippocampal Neurogenesis and the Expression of Autism-Like Behaviors. J Neurosci 2024; 44:e1058232023. [PMID: 38123360 PMCID: PMC10860657 DOI: 10.1523/jneurosci.1058-23.2023] [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: 05/29/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Mutations in the activity-dependent transcription factor MEF2C have been associated with several neuropsychiatric disorders. Among these, autism spectrum disorder (ASD)-related behavioral deficits are manifested. Multiple animal models that harbor mutations in Mef2c have provided compelling evidence that Mef2c is indeed an ASD gene. However, studies in mice with germline or global brain knock-out of Mef2c are limited in their ability to identify the precise neural substrates and cell types that are required for the expression of Mef2c-mediated ASD behaviors. Given the role of hippocampal neurogenesis in cognitive and social behaviors, in this study we aimed to investigate the role of Mef2c in the structure and function of newly generated dentate granule cells (DGCs) in the postnatal hippocampus and to determine whether disrupted Mef2c function is responsible for manifesting ASD behaviors. Overexpression of Mef2c (Mef2cOE ) arrested the transition of neurogenesis at progenitor stages, as indicated by sustained expression of Sox2+ in Mef2cOE DGCs. Conditional knock-out of Mef2c (Mef2ccko ) allowed neuronal commitment of Mef2ccko cells; however, Mef2ccko impaired not only dendritic arborization and spine formation but also synaptic transmission onto Mef2ccko DGCs. Moreover, the abnormal structure and function of Mef2ccko DGCs led to deficits in social interaction and social novelty recognition, which are key characteristics of ASD behaviors. Thus, our study revealed a dose-dependent requirement of Mef2c in the control of distinct steps of neurogenesis, as well as a critical cell-autonomous function of Mef2c in newborn DGCs in the expression of proper social behavior in both sexes.
Collapse
Affiliation(s)
- Sreetama Basu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland 44109, Ohio
| | - Eun Jeoung Ro
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland 44109, Ohio
| | - Zhi Liu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland 44109, Ohio
| | - Hyunjung Kim
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta 30912, Georgia
| | - Aubrey Bennett
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta 30912, Georgia
| | - Seungwoo Kang
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta 30912, Georgia
| | - Hoonkyo Suh
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland 44109, Ohio
| |
Collapse
|
21
|
Zhu F, Shi Q, Jiang YH, Zhang YQ, Zhao H. Impaired synaptic function and hyperexcitability of the pyramidal neurons in the prefrontal cortex of autism-associated Shank3 mutant dogs. Mol Autism 2024; 15:9. [PMID: 38297387 PMCID: PMC10829216 DOI: 10.1186/s13229-024-00587-4] [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: 11/29/2023] [Accepted: 01/22/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND SHANK3 gene is a highly replicated causative gene for autism spectrum disorder and has been well characterized in multiple Shank3 mutant rodent models. When compared to rodents, domestic dogs are excellent animal models in which to study social cognition as they closely interact with humans and exhibit similar social behaviors. Using CRISPR/Cas9 editing, we recently generated a dog model carrying Shank3 mutations, which displayed a spectrum of autism-like behaviors, such as social impairment and heightened anxiety. However, the neural mechanism underlying these abnormal behaviors remains to be identified. METHODS We used Shank3 mutant dog models to examine possible relationships between Shank3 mutations and neuronal dysfunction. We studied electrophysiological properties and the synaptic transmission of pyramidal neurons from acute brain slices of the prefrontal cortex (PFC). We also examined dendrite elaboration and dendritic spine morphology in the PFC using biocytin staining and Golgi staining. We analyzed the postsynaptic density using electron microscopy. RESULTS We established a protocol for the electrophysiological recording of canine brain slices and revealed that excitatory synaptic transmission onto PFC layer 2/3 pyramidal neurons in Shank3 heterozygote dogs was impaired, and this was accompanied by reduced dendrite complexity and spine density when compared to wild-type dogs. Postsynaptic density structures were also impaired in Shank3 mutants; however, pyramidal neurons exhibited hyperexcitability. LIMITATIONS Causal links between impaired PFC pyramidal neuron function and behavioral alterations remain unclear. Further experiments such as manipulating PFC neuronal activity or restoring synaptic transmission in Shank3 mutant dogs are required to assess PFC roles in altered social behaviors. CONCLUSIONS Our study demonstrated the feasibility of using canine brain slices as a model system to study neuronal circuitry and disease. Shank3 haploinsufficiency causes morphological and functional abnormalities in PFC pyramidal neurons, supporting the notion that Shank3 mutant dogs are new and valid animal models for autism research.
Collapse
Affiliation(s)
- Feipeng Zhu
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Shi
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong-Hui Jiang
- Department of Genetics and Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yong Q Zhang
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Life Sciences, Hubei University, Wuhan, 430415, China.
| | - Hui Zhao
- State Key Laboratory for Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
22
|
Dvorak NM, Domingo ND, Tapia CM, Wadsworth PA, Marosi M, Avchalumov Y, Fongsaran C, Koff L, Di Re J, Sampson CM, Baumgartner TJ, Wang P, Villarreal PP, Solomon OD, Stutz SJ, Aditi, Porter J, Gbedande K, Prideaux B, Green TA, Seeley EH, Samir P, Dineley KT, Vargas G, Zhou J, Cisneros I, Stephens R, Laezza F. TNFR1 signaling converging on FGF14 controls neuronal hyperactivity and sickness behavior in experimental cerebral malaria. J Neuroinflammation 2023; 20:306. [PMID: 38115011 PMCID: PMC10729485 DOI: 10.1186/s12974-023-02992-7] [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: 08/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Excess tumor necrosis factor (TNF) is implicated in the pathogenesis of hyperinflammatory experimental cerebral malaria (eCM), including gliosis, increased levels of fibrin(ogen) in the brain, behavioral changes, and mortality. However, the role of TNF in eCM within the brain parenchyma, particularly directly on neurons, remains underdefined. Here, we investigate electrophysiological consequences of eCM on neuronal excitability and cell signaling mechanisms that contribute to observed phenotypes. METHODS The split-luciferase complementation assay (LCA) was used to investigate cell signaling mechanisms downstream of tumor necrosis factor receptor 1 (TNFR1) that could contribute to changes in neuronal excitability in eCM. Whole-cell patch-clamp electrophysiology was performed in brain slices from eCM mice to elucidate consequences of infection on CA1 pyramidal neuron excitability and cell signaling mechanisms that contribute to observed phenotypes. Involvement of identified signaling molecules in mediating behavioral changes and sickness behavior observed in eCM were investigated in vivo using genetic silencing. RESULTS Exploring signaling mechanisms that underlie TNF-induced effects on neuronal excitability, we found that the complex assembly of fibroblast growth factor 14 (FGF14) and the voltage-gated Na+ (Nav) channel 1.6 (Nav1.6) is increased upon tumor necrosis factor receptor 1 (TNFR1) stimulation via Janus Kinase 2 (JAK2). On account of the dependency of hyperinflammatory experimental cerebral malaria (eCM) on TNF, we performed patch-clamp studies in slices from eCM mice and showed that Plasmodium chabaudi infection augments Nav1.6 channel conductance of CA1 pyramidal neurons through the TNFR1-JAK2-FGF14-Nav1.6 signaling network, which leads to hyperexcitability. Hyperexcitability of CA1 pyramidal neurons caused by infection was mitigated via an anti-TNF antibody and genetic silencing of FGF14 in CA1. Furthermore, knockdown of FGF14 in CA1 reduced sickness behavior caused by infection. CONCLUSIONS FGF14 may represent a therapeutic target for mitigating consequences of TNF-mediated neuroinflammation.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Nadia D Domingo
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paul A Wadsworth
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mate Marosi
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yosef Avchalumov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Chanida Fongsaran
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Leandra Koff
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jessica Di Re
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Catherine M Sampson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Timothy J Baumgartner
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Pingyuan Wang
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paula P Villarreal
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Clinical Sciences Program, The Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Olivia D Solomon
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sonja J Stutz
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Aditi
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jacob Porter
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA
| | - Brendan Prideaux
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Erin H Seeley
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Parimal Samir
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kelley T Dineley
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Gracie Vargas
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jia Zhou
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Irma Cisneros
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA.
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
23
|
Proddutur A, Nguyen S, Yeh CW, Gupta A, Santhakumar V. Reclusive chandeliers: Functional isolation of dentate axo-axonic cells after experimental status epilepticus. Prog Neurobiol 2023; 231:102542. [PMID: 37898313 PMCID: PMC10842856 DOI: 10.1016/j.pneurobio.2023.102542] [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: 05/02/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Axo-axonic cells (AACs) provide specialized inhibition to the axon initial segment (AIS) of excitatory neurons and can regulate network output and synchrony. Although hippocampal dentate AACs are structurally altered in epilepsy, physiological analyses of dentate AACs are lacking. We demonstrate that parvalbumin neurons in the dentate molecular layer express PTHLH, an AAC marker, and exhibit morphology characteristic of AACs. Dentate AACs show high-frequency, non-adapting firing but lack persistent firing in the absence of input and have higher rheobase than basket cells suggesting that AACs can respond reliably to network activity. Early after pilocarpine-induced status epilepticus (SE), dentate AACs receive fewer spontaneous excitatory and inhibitory synaptic inputs and have significantly lower maximum firing frequency. Paired recordings and spatially localized optogenetic stimulation revealed that SE reduced the amplitude of unitary synaptic inputs from AACs to granule cells without altering reliability, short-term plasticity, or AIS GABA reversal potential. These changes compromised AAC-dependent shunting of granule cell firing in a multicompartmental model. These early post-SE changes in AAC physiology would limit their ability to receive and respond to input, undermining a critical brake on the dentate throughput during epileptogenesis.
Collapse
Affiliation(s)
- Archana Proddutur
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Susan Nguyen
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Chia-Wei Yeh
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Akshay Gupta
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
24
|
Wenzel M, Huberfeld G, Grayden DB, de Curtis M, Trevelyan AJ. A debate on the neuronal origin of focal seizures. Epilepsia 2023; 64 Suppl 3:S37-S48. [PMID: 37183507 DOI: 10.1111/epi.17650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/26/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023]
Abstract
A critical question regarding how focal seizures start is whether we can identify particular cell classes that drive the pathological process. This was the topic for debate at the recent International Conference for Technology and Analysis of Seizures (ICTALS) meeting (July 2022, Bern, CH) that we summarize here. The debate has been fueled in recent times by the introduction of powerful new ways to manipulate subpopulations of cells in relative isolation, mostly using optogenetics. The motivation for resolving the debate is to identify novel targets for therapeutic interventions through a deeper understanding of the etiology of seizures.
Collapse
Affiliation(s)
- Michael Wenzel
- Department of Epileptology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Gilles Huberfeld
- Neurology Department, Hopital Fondation Adolphe de Rothschild, Paris, France
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - David B Grayden
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- Graeme Clark Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marco de Curtis
- Epilepsy Unit, Fondazione I.R.C.C.S., Istituto Neurologico Carlo Besta, Milan, Italy
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, UK
| |
Collapse
|
25
|
Metcalf CS. Brake Early: RGS14 in CA2 Limits Seizures and Oxidative Stress After SE. Epilepsy Curr 2023; 23:372-374. [PMID: 38269350 PMCID: PMC10805082 DOI: 10.1177/15357597231199343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
RGS14 Limits Seizure-Induced Mitochondrial Oxidative Stress and Pathology in Hippocampus Harbin NH, Lustberg DJ, Hurst C, Pare J, Crotty KM, Waters AL, Yeligar SM, Smith Y, Seyfried NT, Weinschenker D, Hepler JR. Neurobiol Dis. 2023;181:106128. doi:10.1016/j.nbd.2023.106128 . PMID: 37075948 RGS14 is a complex multifunctional scaffolding protein that is highly enriched within pyramidal cells (PCs) of hippocampal area CA2. In these neurons, RGS14 suppresses glutamate-induced calcium influx and related G protein and ERK signaling in dendritic spines to restrain postsynaptic signaling and plasticity. Previous findings show that, unlike PCs of hippocampal areas CA1 and CA3, CA2 PCs are resistant to a number of neurological insults, including degeneration caused by temporal lobe epilepsy (TLE). While RGS14 is protective against peripheral injury, similar roles for RGS14 during pathological injury in hippocampus remain unexplored. Recent studies showed that area CA2 modulates hippocampal excitability, generates epileptiform activity and promotes hippocampal pathology in animal models and patients with TLE. Because RGS14 suppresses CA2 excitability and signaling, we hypothesized that RGS14 would moderate seizure behavior and early hippocampal pathology following seizure activity, possibly affording protection to CA2 PCs. Using kainic acid (KA) to induce status epilepticus (KA-SE) in mice, we show that the loss of RGS14 (RGS14 KO) accelerated onset of limbic motor seizures and mortality compared to wild type (WT) mice, and that KA-SE upregulated RGS14 protein expression in CA2 and CA1 PCs of WT. Our proteomics data show that the loss of RGS14 impacted the expression of a number of proteins at baseline and after KA-SE, many of which associated unexpectedly with mitochondrial function and oxidative stress. RGS14 was shown to localize to the mitochondria in CA2 PCs of mice and reduce mitochondrial respiration in vitro. As a readout of oxidative stress, we found that RGS14 KO dramatically increased 3-nitrotyrosine levels in CA2 PCs, which was greatly exacerbated following KA-SE and correlated with a lack of superoxide dismutase 2 (SOD2) induction. Assessing for hallmarks of seizure pathology in RGS14 KO, we unexpectedly found no differences in neuronal injury in CA2 PCs. However, we observed a striking and surprising lack of microgliosis in CA1 and CA2 of RGS14 KO compared to WT. Together, our data demonstrate a newly appreciated role for RGS14 in limiting intense seizure activity and pathology in hippocampus. Our findings are consistent with a model where RGS14 limits seizure onset and mortality and, after seizure, is upregulated to support mitochondrial function, prevent oxidative stress in CA2 PCs, and promote microglial activation in hippocampus.
Collapse
|
26
|
Whitebirch AC, Santoro B, Barnett A, Lisgaras CP, Scharfman HE, Siegelbaum SA. Reduced Cholecystokinin-Expressing Interneuron Input Contributes to Disinhibition of the Hippocampal CA2 Region in a Mouse Model of Temporal Lobe Epilepsy. J Neurosci 2023; 43:6930-6949. [PMID: 37643861 PMCID: PMC10573827 DOI: 10.1523/jneurosci.2091-22.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023] Open
Abstract
A significant proportion of temporal lobe epilepsy (TLE) patients experience drug-resistant seizures associated with mesial temporal sclerosis, in which there is extensive cell loss in the hippocampal CA1 and CA3 subfields, with a relative sparing of dentate gyrus granule cells and CA2 pyramidal neurons (PNs). A role for CA2 in seizure generation was suggested based on findings of a reduction in CA2 synaptic inhibition (Williamson and Spencer, 1994) and the presence of interictal-like spike activity in CA2 in resected hippocampal tissue from TLE patients (Wittner et al., 2009). We recently found that in the pilocarpine-induced status epilepticus (PILO-SE) mouse model of TLE there was an increase in CA2 intrinsic excitability associated with a loss of CA2 synaptic inhibition. Furthermore, chemogenetic silencing of CA2 significantly reduced seizure frequency, consistent with a role of CA2 in promoting seizure generation and/or propagation (Whitebirch et al., 2022). In the present study, we explored the cellular basis of this inhibitory deficit using immunohistochemical and electrophysiological approaches in PILO-SE male and female mice. We report a widespread decrease in the density of pro-cholecystokinin-immunopositive (CCK+) interneurons and a functional impairment of CCK+ interneuron-mediated inhibition of CA2 PNs. We also found a disruption in the perisomatic perineuronal net in the CA2 stratum pyramidale. Such pathologic alterations may contribute to an enhanced excitation of CA2 PNs and CA2-dependent seizure activity in the PILO-SE mouse model.SIGNIFICANCE STATEMENT Impaired synaptic inhibition in hippocampal circuits has been identified as a key feature that contributes to the emergence and propagation of seizure activity in human patients and animal models of temporal lobe epilepsy (TLE). Among the hippocampal subfields, the CA2 region is particularly resilient to seizure-associated neurodegeneration and has been suggested to play a key role in seizure activity in TLE. Here we report that perisomatic inhibition of CA2 pyramidal neurons mediated by cholecystokinin-expressing interneurons is selectively reduced in acute hippocampal slices from epileptic mice. Parvalbumin-expressing interneurons, in contrast, appear relatively conserved in epileptic mice. These findings advance our understanding of the cellular mechanisms underlying inhibitory disruption in hippocampal circuits in a mouse model of spontaneous recurring seizures.
Collapse
Affiliation(s)
- Alexander C Whitebirch
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University Irving Medical Center, New York, New York 10027
| | - Bina Santoro
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University Irving Medical Center, New York, New York 10027
| | - Anastasia Barnett
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University Irving Medical Center, New York, New York 10027
| | - Christos Panagiotis Lisgaras
- Department of Child & Adolescent Psychiatry, New York University Langone Health, New York, New York 10016
- Department of Neuroscience & Physiology, New York University Langone Health, New York, New York 10016
- Department of Psychiatry, New York University Langone Health, New York, New York 10016
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York 10962
| | - Helen E Scharfman
- Department of Child & Adolescent Psychiatry, New York University Langone Health, New York, New York 10016
- Department of Neuroscience & Physiology, New York University Langone Health, New York, New York 10016
- Department of Psychiatry, New York University Langone Health, New York, New York 10016
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York 10962
| | - Steven A Siegelbaum
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University Irving Medical Center, New York, New York 10027
| |
Collapse
|
27
|
Chameh HM, Falby M, Movahed M, Arbabi K, Rich S, Zhang L, Lefebvre J, Tripathy SJ, De Pittà M, Valiante TA. Distinctive biophysical features of human cell-types: insights from studies of neurosurgically resected brain tissue. Front Synaptic Neurosci 2023; 15:1250834. [PMID: 37860223 PMCID: PMC10584155 DOI: 10.3389/fnsyn.2023.1250834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/21/2023] [Indexed: 10/21/2023] Open
Abstract
Electrophysiological characterization of live human tissue from epilepsy patients has been performed for many decades. Although initially these studies sought to understand the biophysical and synaptic changes associated with human epilepsy, recently, it has become the mainstay for exploring the distinctive biophysical and synaptic features of human cell-types. Both epochs of these human cellular electrophysiological explorations have faced criticism. Early studies revealed that cortical pyramidal neurons obtained from individuals with epilepsy appeared to function "normally" in comparison to neurons from non-epilepsy controls or neurons from other species and thus there was little to gain from the study of human neurons from epilepsy patients. On the other hand, contemporary studies are often questioned for the "normalcy" of the recorded neurons since they are derived from epilepsy patients. In this review, we discuss our current understanding of the distinct biophysical features of human cortical neurons and glia obtained from tissue removed from patients with epilepsy and tumors. We then explore the concept of within cell-type diversity and its loss (i.e., "neural homogenization"). We introduce neural homogenization to help reconcile the epileptogenicity of seemingly "normal" human cortical cells and circuits. We propose that there should be continued efforts to study cortical tissue from epilepsy patients in the quest to understand what makes human cell-types "human".
Collapse
Affiliation(s)
- Homeira Moradi Chameh
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Madeleine Falby
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mandana Movahed
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Scott Rich
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Liang Zhang
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Jérémie Lefebvre
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Mathematics, University of Toronto, Toronto, ON, Canada
| | - Shreejoy J. Tripathy
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Maurizio De Pittà
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Basque Center for Applied Mathematics, Bilbao, Spain
- Faculty of Medicine, University of the Basque Country, Leioa, Spain
| | - Taufik A. Valiante
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, Canada
- Max Planck-University of Toronto Center for Neural Science and Technology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Proddutur A, Nguyen S, Yeh CW, Gupta A, Santhakumar V. RECLUSIVE CHANDELIERS: FUNCTIONAL ISOLATION OF DENTATE AXO-AXONIC CELLS AFTER EXPERIMENTAL STATUS EPILEPTICUS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.01.560378. [PMID: 37873292 PMCID: PMC10592856 DOI: 10.1101/2023.10.01.560378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Axo-axonic cells (AACs) provide specialized inhibition to the axon initial segment (AIS) of excitatory neurons and can regulate network output and synchrony. Although hippocampal dentate AACs are structurally altered in epilepsy, physiological analyses of dentate AACs are lacking. We demonstrate that parvalbumin neurons in the dentate molecular layer express PTHLH, an AAC marker, and exhibit morphology characteristic of AACs. Dentate AACs show high-frequency, non-adapting firing but lack persistent firing in the absence of input and have higher rheobase than basket cells suggesting that AACs can respond reliably to network activity. Early after pilocarpine-induced status epilepticus (SE), dentate AACs receive fewer spontaneous excitatory and inhibitory synaptic inputs and have significantly lower maximum firing frequency. Paired recordings and spatially localized optogenetic stimulation revealed that SE reduced the amplitude of unitary synaptic inputs from AACs to granule cells without altering reliability, short-term plasticity, or AIS GABA reversal potential. These changes compromised AAC-dependent shunting of granule cell firing in a multicompartmental model. These early post-SE changes in AAC physiology would limit their ability to receive and respond to input, undermining a critical brake on the dentate throughput during epileptogenesis.
Collapse
Affiliation(s)
- Archana Proddutur
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Susan Nguyen
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Chia-Wei Yeh
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Akshay Gupta
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| |
Collapse
|
29
|
Takeuchi Y, Yamashiro K, Noguchi A, Liu J, Mitsui S, Ikegaya Y, Matsumoto N. Machine learning-based segmentation of the rodent hippocampal CA2 area from Nissl-stained sections. Front Neuroanat 2023; 17:1172512. [PMID: 37449243 PMCID: PMC10336234 DOI: 10.3389/fnana.2023.1172512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
The hippocampus is a center of learning, memory, and spatial navigation. This region is divided into the CA1, CA2, and CA3 areas, which are anatomically different from each other. Among these divisions, the CA2 area is unique in terms of functional relevance to sociality. The CA2 area is often manually detected based on the size, shape, and density of neurons in the hippocampal pyramidal cell layer, but this manual segmentation relying on cytoarchitecture is impractical to apply to a large number of samples and dependent on experimenters' proficiency. Moreover, the CA2 area has been defined based on expression pattern of molecular marker proteins, but it generally takes days to complete immunostaining for such proteins. Thus, we asked whether the CA2 area can be systematically segmented based on cytoarchitecture alone. Since the expression pattern of regulator of G-protein signaling 14 (RGS14) signifies the CA2 area, we visualized the CA2 area in the mouse hippocampus by RGS14-immunostaining and Nissl-counterstaining and manually delineated the CA2 area. We then established "CAseg," a machine learning-based automated algorithm to segment the CA2 area with the F1-score of approximately 0.8 solely from Nissl-counterstained images that visualized cytoarchitecture. CAseg was extended to the segmentation of the prairie vole CA2 area, which raises the possibility that the use of this algorithm can be expanded to other species. Thus, CAseg will be beneficial for investigating unique properties of the hippocampal CA2 area.
Collapse
Affiliation(s)
- Yuki Takeuchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kotaro Yamashiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Asako Noguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Jiayan Liu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Stöber TM, Batulin D, Triesch J, Narayanan R, Jedlicka P. Degeneracy in epilepsy: multiple routes to hyperexcitable brain circuits and their repair. Commun Biol 2023; 6:479. [PMID: 37137938 PMCID: PMC10156698 DOI: 10.1038/s42003-023-04823-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 04/06/2023] [Indexed: 05/05/2023] Open
Abstract
Due to its complex and multifaceted nature, developing effective treatments for epilepsy is still a major challenge. To deal with this complexity we introduce the concept of degeneracy to the field of epilepsy research: the ability of disparate elements to cause an analogous function or malfunction. Here, we review examples of epilepsy-related degeneracy at multiple levels of brain organisation, ranging from the cellular to the network and systems level. Based on these insights, we outline new multiscale and population modelling approaches to disentangle the complex web of interactions underlying epilepsy and to design personalised multitarget therapies.
Collapse
Affiliation(s)
- Tristan Manfred Stöber
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, 44801, Bochum, Germany
- Epilepsy Center Frankfurt Rhine-Main, Department of Neurology, Goethe University, 60590, Frankfurt, Germany
| | - Danylo Batulin
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
- CePTER - Center for Personalized Translational Epilepsy Research, Goethe University, 60590, Frankfurt, Germany
- Faculty of Computer Science and Mathematics, Goethe University, 60486, Frankfurt, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Peter Jedlicka
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University Giessen, 35390, Giessen, Germany.
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
31
|
Dudek SM, Alexander GM, Farris S. Introduction to the special issue on: A new view of hippocampal area CA2. Hippocampus 2023; 33:127-132. [PMID: 36826426 DOI: 10.1002/hipo.23514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 02/25/2023]
Affiliation(s)
- Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina, USA
| | - Shannon Farris
- Center for Neurobiology Research, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, Virginia, USA
| |
Collapse
|
32
|
Chevaleyre V, Piskorowski R. New hues for CA2. Hippocampus 2023; 33:161-165. [PMID: 36585825 DOI: 10.1002/hipo.23496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 01/01/2023]
Affiliation(s)
- Vivien Chevaleyre
- INSERM U1266, Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, Paris, France.,GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Rebecca Piskorowski
- INSERM U1266, Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, Paris, France.,GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| |
Collapse
|
33
|
Kilias A, Tulke S, Barheier N, Ruther P, Häussler U. Integration of the CA2 region in the hippocampal network during epileptogenesis. Hippocampus 2023; 33:223-240. [PMID: 36421040 DOI: 10.1002/hipo.23479] [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: 08/30/2022] [Revised: 10/14/2022] [Accepted: 11/02/2022] [Indexed: 11/26/2022]
Abstract
The CA2 pyramidal cells are mostly resistant to cell death in mesial temporal lobe epilepsy (MTLE) with hippocampal sclerosis, but they are aberrantly integrated into the epileptic hippocampal network via mossy fiber sprouting. Furthermore, they show increased excitability in vitro in hippocampal slices obtained from human MTLE specimens or animal epilepsy models. Although these changes promote CA2 to contribute to epileptic activity (EA) in vivo, the role of CA2 in the epileptic network within and beyond the sclerotic hippocampus is still unclear. We used the intrahippocampal kainate mouse model for MTLE, which recapitulates most features of the human disease including pharmacoresistant epileptic seizures and hippocampal sclerosis, with preservation of dentate gyrus (DG) granule cells and CA2 pyramidal cells. In vivo recordings with electrodes in CA2 and the DG showed that EA occurs at high coincidence between the ipsilateral DG and CA2 and current source density analysis of silicon probe recordings in dorsal ipsilateral CA2 revealed CA2 as a local source of EA. Cell-specific viral tracing in Amigo2-icreERT2 mice confirmed the preservation of the axonal projection from ipsilateral CA2 pyramidal cells to contralateral CA2 under epileptic conditions and indeed, EA propagated from ipsi- to contralateral CA2 with increasing likelihood with time after KA injection, but always at lower intensity than within the ipsilateral hippocampus. Furthermore, we show that CA2 presents with local theta oscillations and like the DG, shows a pathological reduction of theta frequency already from 2 days after KA onward. The early changes in activity might be facilitated by the loss of glutamic acid decarboxylase 67 (Gad67) mRNA-expressing interneurons directly after the initial status epilepticus in ipsi- but not contralateral CA2. Together, our data highlight CA2 as an active player in the epileptic network and with its contralateral connections as one possible router of aberrant activity.
Collapse
Affiliation(s)
- Antje Kilias
- Institute for Physiology I, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Susanne Tulke
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Nicole Barheier
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Patrick Ruther
- Microsystem Materials Laboratory, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany.,Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Ute Häussler
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| |
Collapse
|
34
|
Harbin NH, Lustberg DJ, Hurst C, Pare JF, Crotty KM, Waters AL, Yeligar SM, Smith Y, Seyfried NT, Weinshenker D, Hepler JR. RGS14 is neuroprotective against seizure-induced mitochondrial oxidative stress and pathology in hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526349. [PMID: 36778349 PMCID: PMC9915580 DOI: 10.1101/2023.02.01.526349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RGS14 is a complex multifunctional scaffolding protein that is highly enriched within pyramidal cells (PCs) of hippocampal area CA2. There, RGS14 suppresses glutamate-induced calcium influx and related G protein and ERK signaling in dendritic spines to restrain postsynaptic signaling and plasticity. Previous findings show that, unlike PCs of hippocampal areas CA1 and CA3, CA2 PCs are resistant to a number of neurological insults, including degeneration caused by temporal lobe epilepsy (TLE). While RGS14 is protective against peripheral injury, similar roles for RGS14 during pathological injury in hippocampus remain unexplored. Recent studies show that area CA2 modulates hippocampal excitability, generates epileptiform activity and promotes hippocampal pathology in animal models and patients with TLE. Because RGS14 suppresses CA2 excitability and signaling, we hypothesized that RGS14 would moderate seizure behavior and early hippocampal pathology following seizure activity. Using kainic acid (KA) to induce status epilepticus (KA-SE) in mice, we show loss of RGS14 (RGS14 KO) accelerated onset of limbic motor seizures and mortality compared to wild type (WT) mice, and that KA-SE upregulated RGS14 protein expression in CA2 and CA1 PCs of WT. Utilizing proteomics, we saw loss of RGS14 impacted the expression of a number of proteins at baseline and after KA-SE, many of which associated unexpectedly with mitochondrial function and oxidative stress. RGS14 was shown to localize to the mitochondria in CA2 PCs of mice and reduce mitochondrial respiration in vitro . As a readout of oxidative stress, we found RGS14 KO dramatically increased 3-nitrotyrosine levels in CA2 PCs, which was greatly exacerbated following KA-SE and correlated with a lack of superoxide dismutase 2 (SOD2) induction. Assessing for hallmarks of seizure pathology in RGS14 KO, we observed worse neuronal injury in area CA3 (but none in CA2 or CA1), and a lack of microgliosis in CA1 and CA2 compared to WT. Together, our data demonstrates a newly appreciated neuroprotective role for RGS14 against intense seizure activity in hippocampus. Our findings are consistent with a model where, after seizure, RGS14 is upregulated to support mitochondrial function and prevent oxidative stress in CA2 PCs, limit seizure onset and hippocampal neuronal injury, and promote microglial activation in hippocampus.
Collapse
|
35
|
Shuman T. Et tu, CA2: CA2 Is Hyperexcitable and Controls Seizures in a Mouse Model of Temporal Lobe Epilepsy. Epilepsy Curr 2023; 23:121-123. [PMID: 37122405 PMCID: PMC10131565 DOI: 10.1177/15357597221150068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
[Box: see text]
Collapse
Affiliation(s)
- Tristan Shuman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai
| |
Collapse
|
36
|
Lisgaras CP, Oliva A, Mckenzie S, LaFrancois J, Siegelbaum SA, Scharman HE. Hippocampal area CA2 controls seizure dynamics, interictal EEG abnormalities and social comorbidity in mouse models of temporal lobe epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524149. [PMID: 36711983 PMCID: PMC9882187 DOI: 10.1101/2023.01.15.524149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Temporal lobe epilepsy (TLE) is characterized by spontaneous recurrent seizures, abnormal activity between seizures, and impaired behavior. CA2 pyramidal neurons (PNs) are potentially important because inhibiting them with a chemogenetic approach reduces seizure frequency in a mouse model of TLE. However, whether seizures could be stopped by timing inhibition just as a seizure begins is unclear. Furthermore, whether inhibition would reduce the cortical and motor manifestations of seizures are not clear. Finally, whether interictal EEG abnormalities and TLE comorbidities would be improved are unknown. Therefore, real-time optogenetic silencing of CA2 PNs during seizures, interictal activity and behavior were studied in 2 mouse models of TLE. CA2 silencing significantly reduced seizure duration and time spent in convulsive behavior. Interictal spikes and high frequency oscillations were significantly reduced, and social behavior was improved. Therefore, brief focal silencing of CA2 PNs reduces seizures, their propagation, and convulsive manifestations, improves interictal EEG, and ameliorates social comorbidities. HIGHLIGHTS Real-time CA2 silencing at the onset of seizures reduces seizure durationWhen CA2 silencing reduces seizure activity in hippocampus it also reduces cortical seizure activity and convulsive manifestations of seizuresInterictal spikes and high frequency oscillations are reduced by real-time CA2 silencingReal-time CA2 silencing of high frequency oscillations (>250Hz) rescues social memory deficits of chronic epileptic mice.
Collapse
|
37
|
Piskorowski RA, Chevaleyre V. Hippocampal area CA2: interneuron disfunction during pathological states. Front Neural Circuits 2023; 17:1181032. [PMID: 37180763 PMCID: PMC10174260 DOI: 10.3389/fncir.2023.1181032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Hippocampal area CA2 plays a critical role in social recognition memory and has unique cellular and molecular properties that distinguish it from areas CA1 and CA3. In addition to having a particularly high density of interneurons, the inhibitory transmission in this region displays two distinct forms of long-term synaptic plasticity. Early studies on human hippocampal tissue have reported unique alteration in area CA2 with several pathologies and psychiatric disorders. In this review, we present recent studies revealing changes in inhibitory transmission and plasticity of area CA2 in mouse models of multiple sclerosis, autism spectrum disorder, Alzheimer's disease, schizophrenia and the 22q11.2 deletion syndrome and propose how these changes could underly deficits in social cognition observed during these pathologies.
Collapse
Affiliation(s)
- Rebecca A. Piskorowski
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
- *Correspondence: Rebecca A. Piskorowski,
| | - Vivien Chevaleyre
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
| |
Collapse
|