1
|
Borjas NC, Anstötz M, Maccaferri G. Multiple layers of diversity govern the cell type specificity of GABAergic input received by mouse subicular pyramidal neurons. J Physiol 2024; 602:4195-4213. [PMID: 39141819 DOI: 10.1113/jp286679] [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: 04/02/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024] Open
Abstract
The subiculum is a key region of the brain involved in the initiation of pathological activity in temporal lobe epilepsy, and local GABAergic inhibition is essential to prevent subicular-originated epileptiform discharges. Subicular pyramidal cells may be easily distinguished into two classes based on their different firing patterns. Here, we have compared the strength of the GABAa receptor-mediated inhibitory postsynaptic currents received by regular- vs. burst-firing subicular neurons and their dynamic modulation by the activation of μ opioid receptors. We have taken advantage of the sequential re-patching of the same cell to initially classify pyramidal neurons according to their firing patters, and then to measure GABAergic events triggered by the optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons. Activation of parvalbumin-expressing cells generated larger responses in postsynaptic burst-firing neurons whereas the opposite was observed for currents evoked by the stimulation of somatostatin-expressing interneurons. In all cases, events depended critically on ω-agatoxin IVA- but not on ω-conotoxin GVIA-sensitive calcium channels. Optogenetic GABAergic input originating from both parvalbumin- and somatostatin-expressing cells was reduced in amplitude following the exposure to a μ opioid receptor agonist. The kinetics of this pharmacological sensitivity was different in regular- vs. burst-firing neurons, but only when responses were evoked by the activation of parvalbumin-expressing neurons, whereas no differences were observed when somatostatin-expressing cells were stimulated. In conclusion, our results show that a high degree of complexity regulates the organizing principles of subicular GABAergic inhibition, with the interaction of pre- and postsynaptic diversity at multiple levels. KEY POINTS: Optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons (PVs and SOMs) triggers inhibitory postsynaptic currents (IPSCs) in both regular- and burst-firing (RFs and BFs) subicular pyramidal cells. The amplitude of optogenetically evoked IPSCs from PVs (PV-opto IPSCs) is larger in BFs whereas IPSCs generated by the light activation of SOMs (SOM-opto IPSCs) are larger in RFs. Both PV- and SOM-opto IPSCs critically depend on ω-agatoxin IVA-sensitive P/Q type voltage-gated calcium channels, whereas no major effects are observed following exposure to ω-conotoxin GVIA, suggesting no significant involvement of N-type channels. The amplitude of both PV- and SOM-opto IPSCs is reduced by the probable pharmacological activation of presynaptic μ opioid receptors, with a faster kinetics of the effect observed in PV-opto IPSCs from RFs vs. BFs, but not in SOM-opto IPSCs. These results help us understand the complex interactions between different layers of diversity regulating GABAergic input onto subicular microcircuits.
Collapse
Affiliation(s)
- Nancy Castro Borjas
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Max Anstötz
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Gianmaria Maccaferri
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, USA
| |
Collapse
|
2
|
Baset A, Huang F. Shedding light on subiculum's role in human brain disorders. Brain Res Bull 2024; 214:110993. [PMID: 38825254 DOI: 10.1016/j.brainresbull.2024.110993] [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: 04/09/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Subiculum is a pivotal output component of the hippocampal formation, a structure often overlooked in neuroscientific research. Here, this review aims to explore the role of the subiculum in various brain disorders, shedding light on its significance within the functional-neuroanatomical perspective on neurological diseases. The subiculum's involvement in multiple brain disorders was thoroughly examined. In Alzheimer's disease, subiculum alterations precede cognitive decline, while in epilepsy, the subiculum plays a critical role in seizure initiation. Stress involves the subiculum's impact on the hypothalamic-pituitary-adrenocortical axis. Moreover, the subiculum exhibits structural and functional changes in anxiety, schizophrenia, and Parkinson's disease, contributing to cognitive deficits. Bipolar disorder is linked to subiculum structural abnormalities, while autism spectrum disorder reveals an alteration of inward deformation in the subiculum. Lastly, frontotemporal dementia shows volumetric differences in the subiculum, emphasizing its contribution to the disorder's complexity. Taken together, this review consolidates existing knowledge on the subiculum's role in brain disorders, and may facilitate future research, diagnostic strategies, and therapeutic interventions for various neurological conditions.
Collapse
Affiliation(s)
- Abdul Baset
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region of China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Special Administrative Region of China
| | - Fengwen Huang
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region of China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Special Administrative Region of China.
| |
Collapse
|
3
|
Griffin WC, Lopez MF, Woodward JJ, Becker HC. Alcohol dependence and the ventral hippocampal influence on alcohol drinking in male mice. Alcohol 2023; 106:44-54. [PMID: 36328184 PMCID: PMC9868110 DOI: 10.1016/j.alcohol.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022]
Abstract
Examining neural circuits underlying persistent, heavy drinking provides insight into the neurobiological mechanisms driving alcohol use disorder. Facilitated by its connectivity with other parts of the brain such as the nucleus accumbens (NAc), the ventral hippocampus (vHC) supports many behaviors, including those related to reward seeking and addiction. These studies used a well-established mouse model of alcohol (ethanol) dependence. After surgery to infuse DREADD-expressing viruses (hM4Di, hM3Dq, or mCherry-only) into the vHC and position guide cannula above the NAc, male C57BL/6J mice were treated in the CIE drinking model that involved repeated cycles of chronic intermittent alcohol (CIE) vapor or air (CTL) exposure alternating with weekly test drinking cycles in which mice were offered alcohol (15% v/v) 2 h/day. Additionally, smaller groups of mice were evaluated for either cFos expression or glutamate release using microdialysis procedures. In CIE mice expressing inhibitory (hM4Di) DREADDs in the vHC, drinking increased as expected, but CNO (3 mg/kg intraperitoneally [i.p.]) given 30 min before testing did not alter alcohol intake. However, in CTL mice expressing hM4Di, CNO significantly increased alcohol drinking (∼30%; p < 0.05) to levels similar to the CIE mice. The vHC-NAc pathway was targeted by infusing CNO into the NAc (3 or 10 μM/side) 30 min before testing. CNO activation of the pathway in mice expressing excitatory (hM3Dq) DREADDs selectively reduced consumption in CIE mice back to CTL levels (∼35-45%; p < 0.05) without affecting CTL alcohol intake. Lastly, activating the vHC-NAc pathway increased cFos expression and evoked significant glutamate release from the vHC terminals in the NAc. These data indicate that reduced activity of the vHC increases alcohol consumption and that targeted, increased activity of the vHC-NAc pathway attenuates excessive drinking associated with alcohol dependence. Thus, these findings indicate that the vHC and its glutamatergic projections to the NAc are involved in excessive alcohol drinking.
Collapse
Affiliation(s)
- William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States.
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States
| | - John J Woodward
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC 29425-0742, United States
| |
Collapse
|
4
|
Zeng C, Lei D, Lu Y, Huang Q, Wu Y, Yang S, Wu Y. Parvalbumin in the metabolic pathway of glutamate and γ-aminobutyric acid: Influence on expression of GAD65 and GAD67. Arch Biochem Biophys 2023; 734:109499. [PMID: 36587827 DOI: 10.1016/j.abb.2022.109499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022]
Abstract
Parvalbumin-expressing neurons are a type of inhibitory intermediate neuron that play an important role in terminating seizures. The aim of the present study was to use lentiviral construction and packaging technology to overexpress and silence the parvalbumin gene in pheochromocytoma (PC12) cells, and to evaluate how parvalbumin influences the metabolic pathway involving glutamate and γ-aminobutyric acid (GABA). In this work, Immunofluorescence staining was used to verify the differentiation of PC12 cells into neurons after adding nerve growth factor (NGF). Western blotting and real-time quantitative polymerase chain reaction (qRT-PCR) were used to confirm lentivirus-mediated knockdown or overexpression of parvalbumin. Expression of parvalbumin, the 65-kDa GAD isoform (GAD65), and the 67-kDa GAD isoform (GAD67) in neuronal cells was examined at the mRNA and protein levels using qRT-PCR, western blotting and immunofluorescence staining, while intracellular glutamate and GABA levels were determined by high performance liquid chromatography (HPLC). We demonstrate that the expression of parvalbumin is associated with GAD65 and GAD67. Interestingly, overexpression of parvalbumin up-regulated GAD65 and GAD67, increased GABA concentration, and decreased glutamate concentration. Silencing of parvalbumin led to the opposite effects. Altogether, parvalbumin affected the expression of GAD65 and GAD67, thereby influencing the metabolic pathway involving glutamate and GABA.
Collapse
Affiliation(s)
- Chunmei Zeng
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Danqing Lei
- Experimental Center of Life Sciences Institutes, Guangxi Medical University, #22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yuling Lu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Qi Huang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Ying Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shengyu Yang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
5
|
Fei F, Wang X, Xu C, Shi J, Gong Y, Cheng H, Lai N, Ruan Y, Ding Y, Wang S, Chen Z, Wang Y. Discrete subicular circuits control generalization of hippocampal seizures. Nat Commun 2022; 13:5010. [PMID: 36008421 PMCID: PMC9411516 DOI: 10.1038/s41467-022-32742-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/15/2022] [Indexed: 11/09/2022] Open
Abstract
Epilepsy is considered a circuit-level dysfunction associated with imbalanced excitation-inhibition, it is therapeutically necessary to identify key brain regions and related circuits in epilepsy. The subiculum is an essential participant in epileptic seizures, but the circuit mechanism underlying its role remains largely elusive. Here we deconstruct the diversity of subicular circuits in a mouse model of epilepsy. We find that excitatory subicular pyramidal neurons heterogeneously control the generalization of hippocampal seizures by projecting to different downstream regions. Notably, anterior thalamus-projecting subicular neurons bidirectionally mediate seizures, while entorhinal cortex-projecting subicular neurons act oppositely in seizure modulation. These two subpopulations are structurally and functionally dissociable. An intrinsically enhanced hyperpolarization-activated current and robust bursting intensity in anterior thalamus-projecting neurons facilitate synaptic transmission, thus contributing to the generalization of hippocampal seizures. These results demonstrate that subicular circuits have diverse roles in epilepsy, suggesting the necessity to precisely target specific subicular circuits for effective treatment of epilepsy.
Collapse
Affiliation(s)
- Fan Fei
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xia Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jiaying Shi
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yiwei Gong
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Heming Cheng
- 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 and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yeping Ruan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Ding
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuang Wang
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. .,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China. .,Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yi Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. .,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China. .,Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Caron D, Canal-Alonso Á, Panuccio G. Mimicking CA3 Temporal Dynamics Controls Limbic Ictogenesis. BIOLOGY 2022; 11:371. [PMID: 35336745 PMCID: PMC8944954 DOI: 10.3390/biology11030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Mesial temporal lobe epilepsy (MTLE) is the most common partial complex epilepsy in adults and the most unresponsive to medications. Electrical deep brain stimulation (DBS) of the hippocampus has proved effective in controlling seizures in epileptic rodents and in drug-refractory MTLE patients. However, current DBS paradigms implement arbitrary fixed-frequency or patterned stimuli, disregarding the temporal profile of brain electrical activity. The latter, herein included hippocampal spontaneous firing, has been shown to follow lognormal temporal dynamics. Here, we present a novel paradigm to devise DBS protocols based on stimulation patterns fashioned as a surrogate brain signal. We focus on the interictal activity originating in the hippocampal subfield CA3, which has been shown to be anti-ictogenic. Using 4-aminopyridine-treated hippocampus-cortex slices coupled to microelectrode array, we pursue three specific aims: (1) address whether lognormal temporal dynamics can describe the CA3-driven interictal pattern, (2) explore the possibility of restoring the non-seizing state by mimicking the temporal dynamics of this anti-ictogenic pattern with electrical stimulation, and (3) compare the performance of the CA3-surrogate against periodic stimulation. We show that the CA3-driven interictal activity follows lognormal temporal dynamics. Further, electrical stimulation fashioned as a surrogate interictal pattern exhibits similar efficacy but uses less pulses than periodic stimulation. Our results support the possibility of mimicking the temporal dynamics of relevant brain signals as a straightforward DBS strategy to ameliorate drug-refractory epilepsy. Further, they herald a paradigm shift in neuromodulation, wherein a compromised brain signal can be recreated by the appropriate stimuli distribution to bypass trial-and-error studies and attain physiologically meaningful DBS operating modes.
Collapse
Affiliation(s)
- Davide Caron
- Enhanced Regenerative Medicine, Istituto Italiano di Tecnologia, 16163 Genova, Italy;
| | - Ángel Canal-Alonso
- BISITE Research Group, University of Salamanca, 37008 Salamanca, Spain;
- Institute for Biomedical Research of Salamanca, University of Salamanca, 37008 Salamanca, Spain
| | - Gabriella Panuccio
- Enhanced Regenerative Medicine, Istituto Italiano di Tecnologia, 16163 Genova, Italy;
| |
Collapse
|
7
|
Antiseizure Effects of Fully Characterized Non-Psychoactive Cannabis sativa L. Extracts in the Repeated 6-Hz Corneal Stimulation Test. Pharmaceuticals (Basel) 2021; 14:ph14121259. [PMID: 34959660 PMCID: PMC8703309 DOI: 10.3390/ph14121259] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Compounds present in Cannabis sativa L. preparations have recently attracted much attention in the treatment of drug-resistant epilepsy. Here, we screened two olive oil extracts from a non-psychoactive C. sativa variety, fully characterized by high-performance liquid chromatography and gas chromatography. Particularly, hemp oils with different concentrations of terpenes were administered at the same dose of cannabidiol (25 mg/kg/day orally), 1 h before the 6-Hz corneal stimulation test (44 mA). Mice were stimulated once a day for 5 days and evaluated by video-electrocorticographic recordings and behavioral analysis. Neuronal activation was assessed by FosB/ΔFosB immunoreactivity. Both oils significantly reduced the percentage of mice experiencing convulsive seizures in comparison to olive oil-treated mice (p < 0.050; Fisher’s exact test), but only the oil enriched with terpenes (K2) significantly accelerated full recovery from the seizure. These effects occurred in the presence of reduced power of delta rhythm, and, instead, increased power of theta rhythm, along with a lower FosB/ΔFosB expression in the subiculum (p < 0.050; Duncan’s method). The overall findings suggest that both cannabinoids and terpenes in oil extracts should be considered as potential therapeutic agents against epileptic seizures and epilepsy.
Collapse
|
8
|
Anstötz M, Fiske MP, Maccaferri G. Impaired KCC2 Function Triggers Interictal-Like Activity Driven by Parvalbumin-Expressing Interneurons in the Isolated Subiculum In Vitro. Cereb Cortex 2021; 31:4681-4698. [PMID: 33987649 PMCID: PMC8408463 DOI: 10.1093/cercor/bhab115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 12/30/2022] Open
Abstract
The decreased expression of the KCC2 membrane transporter in subicular neurons has been proposed to be a key epileptogenic event in temporal lobe epilepsy (TLE). Here, we have addressed this question in a reduced model in vitro and have studied the properties and mechanistic involvement of a major class of interneurons, that is, parvalbumin-expressing cells (PVs). When exposed to the KCC2 blocker VU0463271, mouse subicular slices generated hypersynchronous discharges that could be recorded electrophysiologically and visualized as clusters of co-active neurons with calcium imaging. The pharmacological profile of these events resembled interictal-like discharges in human epileptic tissue because of their dependence on GABAA and AMPA receptors. On average, PVs fired before pyramidal cells (PCs) and the area of co-active clusters was comparable to the individual axonal spread of PVs, suggesting their mechanistic involvement. Optogenetic experiments confirmed this hypothesis, as the flash-stimulation of PVs in the presence of VU0463271 initiated interictal-like discharges, whereas their optogenetic silencing suppressed network hyper-excitability. We conclude that reduced KCC2 activity in subicular networks in vitro is sufficient to induce interictal-like activity via altered GABAergic signaling from PVs without other epilepsy-related changes. This conclusion supports an epileptogenic role for impaired subicular KCC2 function during the progression of TLE.
Collapse
Affiliation(s)
- Max Anstötz
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michael Patrick Fiske
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gianmaria Maccaferri
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
9
|
Synaptic Reshaping and Neuronal Outcomes in the Temporal Lobe Epilepsy. Int J Mol Sci 2021; 22:ijms22083860. [PMID: 33917911 PMCID: PMC8068229 DOI: 10.3390/ijms22083860] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/11/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common types of focal epilepsy, characterized by recurrent spontaneous seizures originating in the temporal lobe(s), with mesial TLE (mTLE) as the worst form of TLE, often associated with hippocampal sclerosis. Abnormal epileptiform discharges are the result, among others, of altered cell-to-cell communication in both chemical and electrical transmissions. Current knowledge about the neurobiology of TLE in human patients emerges from pathological studies of biopsy specimens isolated from the epileptogenic zone or, in a few more recent investigations, from living subjects using positron emission tomography (PET). To overcome limitations related to the use of human tissue, animal models are of great help as they allow the selection of homogeneous samples still presenting a more various scenario of the epileptic syndrome, the presence of a comparable control group, and the availability of a greater amount of tissue for in vitro/ex vivo investigations. This review provides an overview of the structural and functional alterations of synaptic connections in the brain of TLE/mTLE patients and animal models.
Collapse
|
10
|
Fei F, Wang X, Wang Y, Chen Z. Dissecting the role of subiculum in epilepsy: Research update and translational potential. Prog Neurobiol 2021; 201:102029. [PMID: 33636224 DOI: 10.1016/j.pneurobio.2021.102029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 01/12/2021] [Accepted: 02/21/2021] [Indexed: 11/25/2022]
Abstract
The subiculum serves as the strategic core output of the hippocampus, through which neural activity exits the hippocampal proper and targets the entorhinal cortex and other more distant subcortical and cortical areas. The past decade has witnessed a growing interest in the subiculum, owing to discoveries revealing its critical role in regulating many physiological and pathophysiological processes. Notably, accumulating evidence from both clinical and experimental studies suggests that the subiculum plays a vital role in seizure initiation and propagation, in epilepsy. In this review, we briefly describe the structure and connectivity of the subiculum and then summarize the molecular and cellular mechanisms in the subiculum underlying the epileptic brain, in both epilepsy patients and animal models. Next, we review some translational approaches targeting the malfunctioned subiculum to treat epilepsy. Finally, we pose open questions for future research in the subiculum and their clinical translation challenges.
Collapse
Affiliation(s)
- Fan Fei
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xia Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China; Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
Lévesque M, Biagini G, Avoli M. Neurosteroids and Focal Epileptic Disorders. Int J Mol Sci 2020; 21:ijms21249391. [PMID: 33321734 PMCID: PMC7763947 DOI: 10.3390/ijms21249391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/27/2020] [Accepted: 12/08/2020] [Indexed: 11/18/2022] Open
Abstract
Neurosteroids are a family of compounds that are synthesized in principal excitatory neurons and glial cells, and derive from the transformation of cholesterol into pregnenolone. The most studied neurosteroids—allopregnanolone and allotetrahydrodeoxycorticosterone (THDOC)—are known to modulate GABAA receptor-mediated transmission, thus playing a role in controlling neuronal network excitability. Given the role of GABAA signaling in epileptic disorders, neurosteroids have profound effects on seizure generation and play a role in the development of chronic epileptic conditions (i.e., epileptogenesis). We review here studies showing the effects induced by neurosteroids on epileptiform synchronization in in vitro brain slices, on epileptic activity in in vivo models, i.e., in animals that were made epileptic with chemoconvulsant treatment, and in epileptic patients. These studies reveal that neurosteroids can modulate ictogenesis and the occurrence of pathological network activity such as interictal spikes and high-frequency oscillations (80–500 Hz). Moreover, they can delay the onset of spontaneous seizures in animal models of mesial temporal lobe epilepsy. Overall, this evidence suggests that neurosteroids represent a new target for the treatment of focal epileptic disorders.
Collapse
Affiliation(s)
- Maxime Lévesque
- Montreal Neurological Institute-Hospital & Department of Neurology and Neurosurgery, 3801 University Street, Montreal, QC H3A 2B4, Canada;
- Correspondence: ; Tel.: +1-514-398-8909
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Università 4, 41121 Modena, Italy;
| | - Massimo Avoli
- Montreal Neurological Institute-Hospital & Department of Neurology and Neurosurgery, 3801 University Street, Montreal, QC H3A 2B4, Canada;
- Department of Physiology, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
12
|
Lévesque M, Avoli M. The subiculum and its role in focal epileptic disorders. Rev Neurosci 2020; 32:249-273. [PMID: 33661586 DOI: 10.1515/revneuro-2020-0091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/29/2020] [Indexed: 01/07/2023]
Abstract
The subicular complex (hereafter referred as subiculum), which is reciprocally connected with the hippocampus and rhinal cortices, exerts a major control on hippocampal outputs. Over the last three decades, several studies have revealed that the subiculum plays a pivotal role in learning and memory but also in pathological conditions such as mesial temporal lobe epilepsy (MTLE). Indeed, subicular networks actively contribute to seizure generation and this structure is relatively spared from the cell loss encountered in this focal epileptic disorder. In this review, we will address: (i) the functional properties of subicular principal cells under normal and pathological conditions; (ii) the subiculum role in sustaining seizures in in vivo models of MTLE and in in vitro models of epileptiform synchronization; (iii) its presumptive role in human MTLE; and (iv) evidence underscoring the relationship between subiculum and antiepileptic drug effects. The studies reviewed here reinforce the view that the subiculum represents a limbic area with relevant, as yet unexplored, roles in focal epilepsy.
Collapse
Affiliation(s)
- Maxime Lévesque
- Departments of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, H3A 2B4Québec, Canada
| | - Massimo Avoli
- Departments of Neurology, Neurosurgery, and Physiology, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, H3A 2B4Québec, Canada
| |
Collapse
|
13
|
Duy PQ, He M, He Z, Kahle KT. Preclinical insights into therapeutic targeting of KCC2 for disorders of neuronal hyperexcitability. Expert Opin Ther Targets 2020; 24:629-637. [PMID: 32336175 DOI: 10.1080/14728222.2020.1762174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Epilepsy is a common neurological disorder of neuronal hyperexcitability that begets recurrent and unprovoked seizures. The lack of a truly satisfactory pharmacotherapy for epilepsy highlights the clinical urgency for the discovery of new drug targets. To that end, targeting the electroneutral K+/Cl- cotransporter KCC2 has emerged as a novel therapeutic strategy for the treatment of epilepsy. AREAS COVERED We summarize the roles of KCC2 in the maintenance of synaptic inhibition and the evidence linking KCC2 dysfunction to epileptogenesis. We also discuss preclinical proof-of-principle studies that demonstrate that augmentation of KCC2 function can reduce seizure activity. Moreover, potential strategies to modulate KCC2 activity for therapeutic benefit are highlighted. EXPERT OPINION Although KCC2 is a promising drug target, questions remain before clinical translation. It is unclear whether increasing KCC2 activity can reverse epileptogenesis, the ultimate curative goal for epilepsy therapy that extends beyond seizure reduction. Furthermore, the potential adverse effects associated with increased KCC2 function have not been studied. Continued investigations into the neurobiology of KCC2 will help to translate promising preclinical insights into viable therapeutic avenues that leverage fundamental properties of KCC2 to treat medically intractable epilepsy and other disorders of failed synaptic inhibition with attendant neuronal hyperexcitability.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Medical Scientist Training Program, Yale University School of Medicine , New Haven, CT, USA
| | - Miao He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Department of Genetics, Yale University School of Medicine , New Haven, CT, USA.,Departments of Pediatrics and Cellular & Molecular Physiology, Yale University School of Medicine , New Haven, CT, USA.,Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale University , New Haven, CT, USA.,Yale Stem Cell Center, Yale School of Medicine , New Haven, CT, USA
| |
Collapse
|
14
|
Low-frequency Stimulation at the Subiculum is Anti-convulsant and Anti-drug-resistant in a Mouse Model of Lamotrigine-resistant Temporal Lobe Epilepsy. Neurosci Bull 2020; 36:654-658. [PMID: 32157502 DOI: 10.1007/s12264-020-00482-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/10/2019] [Indexed: 10/24/2022] Open
|
15
|
Auer T, Schreppel P, Erker T, Schwarzer C. Impaired chloride homeostasis in epilepsy: Molecular basis, impact on treatment, and current treatment approaches. Pharmacol Ther 2020; 205:107422. [DOI: 10.1016/j.pharmthera.2019.107422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
|
16
|
Wengert ER, Saga AU, Panchal PS, Barker BS, Patel MK. Prax330 reduces persistent and resurgent sodium channel currents and neuronal hyperexcitability of subiculum neurons in a mouse model of SCN8A epileptic encephalopathy. Neuropharmacology 2019; 158:107699. [PMID: 31278928 DOI: 10.1016/j.neuropharm.2019.107699] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/25/2019] [Accepted: 07/01/2019] [Indexed: 11/28/2022]
Abstract
SCN8A epileptic encephalopathy is a severe genetic epilepsy syndrome caused by de novo gain-of-function mutations of SCN8A encoding the voltage-gated sodium (Na) channel (VGSC) NaV1.6. Therapeutic management is difficult in many patients, leading to uncontrolled seizures and risk of sudden unexpected death in epilepsy (SUDEP). There is a need to develop novel anticonvulsants that can specifically target aberrant VGSC activity associated with SCN8A gain-of-function mutations. In this study, we investigate the effects of Prax330, a novel VGSC inhibitor, on the biophysical properties of wild-type (WT) NaV1.6 and the patient mutation p.Asn1768Asp (N1768D) in ND7/23 cells. The effects of Prax330 on persistent (INaP) and resurgent (INaR) Na currents and neuronal excitability in subiculum neurons from a knock-in mouse model of the Scn8a-N1768D mutation (Scn8aD/+) were also examined. In ND7/23 cells, Prax330 reduced INaP currents recorded from cells expressing Scn8a-N1768D and hyperpolarized steady-state inactivation curves. Recordings from brain slices demonstrated elevated INaP and INaR in subiculum neurons from Scn8aD/+ mutant mice and abnormally large action potential (AP) burst-firing events in a subset of neurons. Prax330 (1 μM) reduced both INaP and INaR and suppressed AP bursts, with a smaller effect on AP waveforms that had similar morphology to WT neurons. Prax330 (1 μM) also reduced synaptically-evoked APs in Scn8aD/+ subiculum neurons but not in WT neurons. Our results highlight the efficacy of targeting INaP and INaR and inactivation parameters in controlling subiculum excitability and suggest Prax330 as a promising novel therapy for SCN8A epileptic encephalopathy.
Collapse
Affiliation(s)
- Eric R Wengert
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908, USA; Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Anusha U Saga
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Payal S Panchal
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Bryan S Barker
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908, USA; Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Manoj K Patel
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, 22908, USA; Neuroscience Graduate Program, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
17
|
Wang Y, Wang Y, Chen Z. Double-edged GABAergic synaptic transmission in seizures: The importance of chloride plasticity. Brain Res 2018; 1701:126-136. [PMID: 30201259 DOI: 10.1016/j.brainres.2018.09.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
GABAergic synaptic inhibition, which is a critical regulator of neuronal excitability, is closely involved in epilepsy. Interestingly, fast GABAergic transmission mediated by Cl- permeable GABAA receptors can bi-directionally exert both seizure-suppressing and seizure-promoting actions. Accumulating evidence suggests that chloride plasticity, the driving force of GABAA receptor-mediated synaptic transmission, contributes to the double-edged role of GABAergic synapses in seizures. Large amounts of Cl- influx can overwhelm Cl- extrusion during seizures not only in healthy tissue in a short-term "activity-dependent" manner, but also in chronic epilepsy in a long-term, irreversible "pathology-dependent" manner related to the dysfunction of two chloride transporters: the chloride importer NKCC1 and the chloride exporter KCC2. In this review, we address the importance of chloride plasticity for the "activity-dependent" and "pathology-dependent" mechanisms underlying epileptic events and provide possible directions for further research, which may be clinically important for the design of GABAergic synapse-targeted precise therapeutic interventions for epilepsy.
Collapse
Affiliation(s)
- Ying Wang
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
18
|
Zhang L, Fan D, Wang Q, Baier G. Effects of brain-derived neurotrophic factor and noise on transitions in temporal lobe epilepsy in a hippocampal network. CHAOS (WOODBURY, N.Y.) 2018; 28:106322. [PMID: 30384669 DOI: 10.1063/1.5036690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/19/2018] [Indexed: 06/08/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has recently been implicated in the modulation of receptor activation leading to dynamic state transitions in temporal lobe epilepsy (TLE). In addition, the crucial role of neuronal noise in these transitions has been studied in electrophysiological experiments. However, the precise role of these factors during seizure generation in TLE is not known. Building on a previously proposed model of an epileptogenic hippocampal network, we included the actions of BDNF-regulated receptors and intrinsic noise. We found that the effects of both BDNF and noise can increase the activation of N-methyl-D-aspartate receptors leading to excessive C a 2 + flux, which induces abnormal fast spiking and bursting. Our results indicate that the combined effects have a strong influence on the seizure-generating network, resulting in higher firing frequency and amplitude. As correlations between firing increase, the synchronization of the entire network increases, a marker of the ictogenic transitions from normal to seizures-like dynamics. Our work on the effects of BDNF dynamics in a noisy environment might lead to an improved model-based understanding of the pathological mechanisms in TLE.
Collapse
Affiliation(s)
- Liyuan Zhang
- Department of Dynamics and Control, Beihang University, 100191 Beijing, China
| | - Denggui Fan
- School of Mathematics and Physics, University of Science and Technology Beijing, 100083 Beijing, China
| | - Qingyun Wang
- Department of Dynamics and Control, Beihang University, 100191 Beijing, China
| | - Gerold Baier
- Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
19
|
Voltage-Dependent Calcium Channels, Calcium Binding Proteins, and Their Interaction in the Pathological Process of Epilepsy. Int J Mol Sci 2018; 19:ijms19092735. [PMID: 30213136 PMCID: PMC6164075 DOI: 10.3390/ijms19092735] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023] Open
Abstract
As an important second messenger, the calcium ion (Ca2+) plays a vital role in normal brain function and in the pathophysiological process of different neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and epilepsy. Ca2+ takes part in the regulation of neuronal excitability, and the imbalance of intracellular Ca2+ is a trigger factor for the occurrence of epilepsy. Several anti-epileptic drugs target voltage-dependent calcium channels (VDCCs). Intracellular Ca2+ levels are mainly controlled by VDCCs located in the plasma membrane, the calcium-binding proteins (CBPs) inside the cytoplasm, calcium channels located on the intracellular calcium store (particular the endoplasmic reticulum/sarcoplasmic reticulum), and the Ca2+-pumps located in the plasma membrane and intracellular calcium store. So far, while many studies have established the relationship between calcium control factors and epilepsy, the mechanism of various Ca2+ regulatory factors in epileptogenesis is still unknown. In this paper, we reviewed the function, distribution, and alteration of VDCCs and CBPs in the central nervous system in the pathological process of epilepsy. The interaction of VDCCs with CBPs in the pathological process of epilepsy was also summarized. We hope this review can provide some clues for better understanding the mechanism of epileptogenesis, and for the development of new anti-epileptic drugs targeting on VDCCs and CBPs.
Collapse
|
20
|
Di Cristo G, Awad PN, Hamidi S, Avoli M. KCC2, epileptiform synchronization, and epileptic disorders. Prog Neurobiol 2018; 162:1-16. [DOI: 10.1016/j.pneurobio.2017.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/28/2017] [Indexed: 12/31/2022]
|
21
|
Ye H, Kaszuba S. Inhibitory or excitatory? Optogenetic interrogation of the functional roles of GABAergic interneurons in epileptogenesis. J Biomed Sci 2017; 24:93. [PMID: 29202749 PMCID: PMC5715558 DOI: 10.1186/s12929-017-0399-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/28/2017] [Indexed: 01/22/2023] Open
Abstract
Alteration in the excitatory/inhibitory neuronal balance is believed to be the underlying mechanism of epileptogenesis. Based on this theory, GABAergic interneurons are regarded as the primary inhibitory neurons, whose failure of action permits hyperactivity in the epileptic circuitry. As a consequence, optogenetic excitation of GABAergic interneurons is widely used for seizure suppression. However, recent evidence argues for the context-dependent, possibly “excitatory” roles that GABAergic cells play in epileptic circuitry. We reviewed current optogenetic approaches that target the “inhibitory” roles of GABAergic interneurons for seizure control. We also reviewed interesting evidence that supports the “excitatory” roles of GABAergic interneurons in epileptogenesis. GABAergic interneurons can provide excitatory effects to the epileptic circuits via several distinct neurological mechanisms. (1) GABAergic interneurons can excite postsynaptic neurons, due to the raised reversal potential of GABA receptors in the postsynaptic cells. (2) Continuous activity in GABAergic interneurons could lead to transient GABA depletion, which prevents their inhibitory effect on pyramidal cells. (3) GABAergic interneurons can synchronize network activity during seizure. (4) Some GABAergic interneurons inhibit other interneurons, causing disinhibition of pyramidal neurons and network hyperexcitability. The dynamic, context-dependent role that GABAergic interneurons play in seizure requires further investigation of their functions at single cell and circuitry level. New optogenetic protocols that target GABAergic inhibition should be explored for seizure suppression.
Collapse
Affiliation(s)
- Hui Ye
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd, Chicago, IL, 60660, USA.
| | - Stephanie Kaszuba
- Department of Biology, Loyola University Chicago, Quinlan Life Sciences Education and Research Center, 1032 W. Sheridan Rd, Chicago, IL, 60660, USA
| |
Collapse
|
22
|
Pro-excitatory alterations in sodium channel activity facilitate subiculum neuron hyperexcitability in temporal lobe epilepsy. Neurobiol Dis 2017; 108:183-194. [PMID: 28860087 DOI: 10.1016/j.nbd.2017.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/07/2017] [Accepted: 08/26/2017] [Indexed: 11/23/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is a common form of adult epilepsy involving the limbic structures of the temporal lobe. Subiculum neurons act to provide a major output from the hippocampus and consist of a large population of endogenously bursting excitatory neurons. In TLE, subiculum neurons are largely spared, become hyperexcitable and show spontaneous epileptiform activity. The basis for this hyperexcitability is unclear, but is likely to involve alterations in the expression levels and function of various ion channels. In this study, we sought to determine the importance of sodium channel currents in facilitating neuronal hyperexcitability of subiculum neurons in the continuous hippocampal stimulation (CHS) rat model of TLE. Subiculum neurons from TLE rats were hyperexcitable, firing a higher frequency of action potentials after somatic current injection and action potential (AP) bursts after synaptic stimulation. Voltage clamp recordings revealed increases in resurgent (INaR) and persistent (INaP) sodium channel currents and pro-excitatory shifts in sodium channel activation and inactivation parameters that would facilitate increases in AP generation. Attenuation of INaR and INaP currents with 4,9-anhydro-tetrodotoxin (4,9-ah TTX; 100nM), a toxin with increased potency against Nav1.6 channels, suppressed neuronal firing frequency and inhibited AP bursting induced by synaptic stimulation in TLE neurons. These findings support an important role of sodium channels, particularly Nav1.6, in facilitating subiculum neuron hyperexcitability in TLE and provide further support for the importance of INaR and INaP currents in establishing epileptiform activity of subiculum neurons.
Collapse
|
23
|
Allopregnanolone decreases interictal spiking and fast ripples in an animal model of mesial temporal lobe epilepsy. Neuropharmacology 2017; 121:12-19. [DOI: 10.1016/j.neuropharm.2017.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 04/13/2017] [Indexed: 11/21/2022]
|
24
|
Depolarized GABAergic Signaling in Subicular Microcircuits Mediates Generalized Seizure in Temporal Lobe Epilepsy. Neuron 2017. [PMID: 28648501 DOI: 10.1016/j.neuron.2017.06.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Secondary generalized seizure (sGS) is a major source of disability in temporal lobe epilepsy (TLE) with unclear cellular/circuit mechanisms. Here we found that clinical TLE patients with sGS showed reduced volume specifically in the subiculum compared with those without sGS. Further, using optogenetics and extracellular electrophysiological recording in mouse models, we found that photoactivation of subicular GABAergic neurons retarded sGS acquisition by inhibiting the firing of pyramidal neurons. Once sGS had been stably acquired, photoactivation of GABAergic neurons aggravated sGS expression via depolarized GABAergic signaling. Subicular parvalbumin, but not somatostatin subtype GABAergic, neurons were easily depolarized in sGS expression. Finally, photostimulation of subicular pyramidal neurons genetically targeted with proton pump Arch, rather than chloride pump NpHR3.0, alleviated sGS expression. These results demonstrated that depolarized GABAergic signaling in subicular microcircuit mediates sGS in TLE. This may be of therapeutic interest in understanding the pathological neuronal circuitry underlying sGS. VIDEO ABSTRACT.
Collapse
|
25
|
Kelley MR, Deeb TZ, Brandon NJ, Dunlop J, Davies PA, Moss SJ. Compromising KCC2 transporter activity enhances the development of continuous seizure activity. Neuropharmacology 2016; 108:103-10. [PMID: 27108931 PMCID: PMC5337122 DOI: 10.1016/j.neuropharm.2016.04.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/05/2016] [Accepted: 04/20/2016] [Indexed: 11/17/2022]
Abstract
Impaired neuronal inhibition has long been associated with the increased probability of seizure occurrence and heightened seizure severity. Fast synaptic inhibition in the brain is primarily mediated by the type A γ-aminobutyric acid receptors (GABAARs), ligand-gated ion channels that can mediate Cl(-) influx resulting in membrane hyperpolarization and the restriction of neuronal firing. In most adult brain neurons, the K(+)/Cl(-) co-transporter-2 (KCC2) establishes hyperpolarizing GABAergic inhibition by maintaining low [Cl(-)]i. In this study, we sought to understand how decreased KCC2 transport function affects seizure event severity. We impaired KCC2 transport in the 0-Mg(2+) ACSF and 4-aminopyridine in vitro models of epileptiform activity in acute mouse brain slices. Experiments with the selective KCC2 inhibitor VU0463271 demonstrated that reduced KCC2 transport increased the duration of SLEs, resulting in non-terminating discharges of clonic-like activity. We also investigated slices obtained from the KCC2-Ser940Ala (S940A) point-mutant mouse, which has a mutation at a known functional phosphorylation site causing behavioral and cellular deficits under hyperexcitable conditions. We recorded from the entorhinal cortex of S940A mouse brain slices in both 0-Mg(2+) ACSF and 4-aminopyridine, and demonstrated that loss of the S940 residue increased the susceptibility of continuous clonic-like discharges, an in vitro form of status epilepticus. Our experiments revealed KCC2 transport activity is a critical factor in seizure event duration and mechanisms of termination. Our results highlight the need for therapeutic strategies that potentiate KCC2 transport function in order to decrease seizure event severity and prevent the development of status epilepticus.
Collapse
Affiliation(s)
- Matthew R Kelley
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Tarek Z Deeb
- AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA
| | - Nicholas J Brandon
- AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, 141, Portland St, Cambridge, MA, USA
| | - John Dunlop
- AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, 141, Portland St, Cambridge, MA, USA
| | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA; AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Boston, MA 02111, USA; Department of Neuroscience, Physiology and Pharmacology, University College, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Jaggi AS, Kaur A, Bali A, Singh N. Expanding Spectrum of Sodium Potassium Chloride Co-transporters in the Pathophysiology of Diseases. Curr Neuropharmacol 2016; 13:369-88. [PMID: 26411965 PMCID: PMC4812803 DOI: 10.2174/1570159x13666150205130359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Sodium potassium chloride co-transporter (NKCC) belongs to cation-dependent chloride co-transporter family, whose activation allows the entry of Na(+), K(+) and 2Cl(-) inside the cell. It acts in concert with K(+) Cl(-) co-transporter (KCC), which extrudes K(+) and Cl(-) ions from cell. NKCC1 is widely distributed throughout the body, while NKCC2 is exclusively present in kidney. Protein kinase A, protein kinase C, Ste20-related proline-alanine-rich kinase, oxidative stress responsive kinases, With No K=lysine kinase and protein phosphatase type 1 control the phosphorylation/dephosphorylation of key threonine residues of in regulatory domain of NKCC1. The selective inhibitors of NKCC1 including bumetanide and furosemide are conventionally employed as diuretics. However, recent studies have indicated that NKCC1 may be involved in the pathophysiology of anxiety, cerebral ischemia, epilepsy, neuropathic pain, fragile X syndrome, autism and schizophrenia. The inhibitors of NKCC1 are shown to produce anxiolytic effects; attenuate cerebral ischemia-induced neuronal injury; produce antiepileptic effects and attenuate neuropathic pain. In the early developing brain, GABAA activation primarily produces excitatory actions due to high NKCC1/KCC2 ratio. However, as the development progresses, the ratio of NKCC1/KCC2 ratio reverses and there is switch in the polarity of GABAA actions and latter acquires the inhibitory actions. The recapitulation of developmental-like state during pathological state may be associated with increase in the expression and functioning of NKCC1, which decreases the strength of inhibitory GABAergic neurotransmission. The present review describes the expanding role and mechanism of NKCC1 in the pathophysiology of different diseases.
Collapse
Affiliation(s)
- Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala- 147002.
| | | | | | | |
Collapse
|
27
|
Developmental pharmacology of benzodiazepines under normal and pathological conditions. Epileptic Disord 2016; 16 Spec No 1:S59-68. [PMID: 25335485 DOI: 10.1684/epd.2014.0690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Benzodiazepines are allosteric agonists of GABAA receptors (GABAAR), pentameric ligand-gated Cl(-) channels, which serve both an important neurodevelopmental role but are also the principal inhibitory system in the brain. However, their subunit composition, channel properties, and function, as well as their region-specific expression patterns, change through development. These processes have been extensively studied in rodents and to some extent confirmed in higher species. Specifically, GABAARs acquire faster kinetics with age and their pharmacology changes rendering them more sensitive to drugs that have higher affinity for α1 subunit-containing GABAARs, such as benzodiazepines, but also, their inhibitory function becomes more potent as they shift from having depolarising to hyperpolarising responses due to a shift in Cl(-) gradient and cation chloride cotransporter expression. Concerns have been raised about possible pro-apoptotic and paradoxical effects of benzodiazepines in the neonatal normal rat brain, although it is unclear, as yet, whether this extends to brains exposed to seizures. Growing evidence indicates that the pharmacology and physiology of GABAARs may be altered in the brain of rats or humans with seizures or epilepsy, or different aetiologies that predispose to epilepsy. These changes follow different paths, depending on sex, age, region, cell type, aetiology, or time-point specific factors. Identification of dynamic biomarkers that could enable these changes in vivo to be monitored would greatly facilitate the selection of more effective agonists with fewer side effects.
Collapse
|
28
|
Behr C, Lévesque M, Ragsdale D, Avoli M. Lacosamide modulates interictal spiking and high-frequency oscillations in a model of mesial temporal lobe epilepsy. Epilepsy Res 2015; 115:8-16. [PMID: 26220372 PMCID: PMC4878889 DOI: 10.1016/j.eplepsyres.2015.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/28/2015] [Accepted: 05/12/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Nearly one third of patients presenting with mesial temporal lobe epilepsy (MTLE), the most prevalent lesion-related epileptic disorder in adulthood, do not respond to currently available antiepileptic medications. Thus, there is a need to identify and characterize new antiepileptic drugs. In this study, we used the pilocarpine model of MTLE to establish the effects of a third generation drug, lacosamide (LCM), on seizures, interictal spikes and high-frequency oscillations (HFOs, ripples: 80-200 Hz, fast ripples: 250-500 Hz). METHODS Sprague-Dawley rats (250-300 g) were injected with pilocarpine to induce a status epilepticus (SE) that was pharmacologically terminated after 1h. Eight pilocarpine-treated rats were then injected with LCM (30 mg/kg, i.p.) 4h after SE and daily for 14 days. Eight pilocarpine-treated rats were used as controls and treated with saline. Three days after SE, all rats were implanted with bipolar electrodes in the hippocampal CA3 region, entorhinal cortex (EC), dentate gyrus (DG) and subiculum and EEG-video monitored from day 4 to day 14 after SE. RESULTS LCM-treated animals showed lower rates of seizures (0.21 (± 0.11) seizures/day) than controls (2.6 (±0.57), p<0.05), and a longer latent period (LCM: 11 (± 1) days, controls: 6.25 (± 1), p<0.05). Rates of interictal spikes in LCM-treated rats were significantly lower than in controls in CA3 and subiculum (p<0.05). Rates of ripples and fast ripples associated with interictal spikes in CA3 and subiculum as well as rates of fast ripples occurring outside of interictal spikes in CA3 were also significantly lower in LCM-treated animals. In controls, interictal spikes and associated HFOs correlated to seizure clustering, while this was not the case for isolated HFOs. SIGNIFICANCE Our findings show that early treatment with LCM has powerful anti-ictogenic properties in the pilocarpine model of MTLE. These effects are accompanied by decreased rates of interictal spikes and associated HFOs. Isolated HFOs were also modulated by LCM, in a manner that appeared to be unrelated to its antiictogenic effects. These results thus suggest that distinct mechanisms may underlie interictal-associated and isolated HFOs in the pilocarpine model of MTLE.
Collapse
Affiliation(s)
- Charles Behr
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montréal, QC, Canada H3A 2B4
| | - Maxime Lévesque
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montréal, QC, Canada H3A 2B4
| | - David Ragsdale
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montréal, QC, Canada H3A 2B4
| | - Massimo Avoli
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montréal, QC, Canada H3A 2B4.
| |
Collapse
|
29
|
Dhaher R, Wang H, Gruenbaum SE, Tu N, Lee TSW, Zaveri HP, Eid T. Effects of site-specific infusions of methionine sulfoximine on the temporal progression of seizures in a rat model of mesial temporal lobe epilepsy. Epilepsy Res 2015. [PMID: 26220375 DOI: 10.1016/j.eplepsyres.2015.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glutamine synthetase (GS) in astrocytes is critical for metabolism of glutamate and ammonia in the brain, and perturbations in the anatomical distribution and activity of the enzyme are likely to adversely affect synaptic transmission. GS is deficient in discrete regions of the hippocampal formation in patients with mesial temporal lobe epilepsy (MTLE), a disorder characterized by brain glutamate excess and recurrent seizures. To investigate the role of site-specific inhibition of GS in MTLE, we chronically infused the GS inhibitor methionine sulfoximine (MSO) into one of the following areas of adult laboratory rats: (1) the angular bundle, n=6; (2) the deep entorhinal cortex (EC), n=7; (3) the stratum lacunosum-moleculare of CA1, n=7; (4) the molecular layer of the subiculum, n=10; (5) the hilus of the dentate gyrus, n=6; and (6) the lateral ventricle, n=6. Twelve animals were infused with phosphate buffered saline (PBS) into the same areas to serve as controls. All infusions were unilateral, and animals were monitored by continuous video-intracranial EEG recordings for 3 weeks to capture seizure activity. All animals infused with MSO into the entorhinal-hippocampal area exhibited recurrent seizures that were particularly frequent during the first 3 days of infusion and that continued to recur for the entire 3 week recording period. Only a fraction of animals infused with MSO into the lateral ventricle had recurrent seizures, which occurred at a lower frequency compared with the other MSO infused group. Infusion of MSO into the hilus of the dentate gyrus resulted in the highest total number of seizures over the 3-week recording period. Infusion of MSO into all brain regions studied, with the exception of the lateral ventricle, led to a change in the composition of seizure severity over time. Low-grade (stages 1-3) seizures were more prevalent early during infusion, while severe (stages 4-5) seizures were more prevalent later. Thus, the site of GS inhibition within the brain determines the pattern and temporal evolution of recurrent seizures in the MSO model of MTLE.
Collapse
Affiliation(s)
- Roni Dhaher
- Department of Laboratory Medicine, Yale School of Medicine, 330 Cedar St., PO Box 208083, New Haven, CT 06520-8035, USA
| | - Helen Wang
- Department of Laboratory Medicine, Yale School of Medicine, 330 Cedar St., PO Box 208083, New Haven, CT 06520-8035, USA
| | - Shaun E Gruenbaum
- Department of Laboratory Medicine, Yale School of Medicine, 330 Cedar St., PO Box 208083, New Haven, CT 06520-8035, USA
| | - Nathan Tu
- Department of Laboratory Medicine, Yale School of Medicine, 330 Cedar St., PO Box 208083, New Haven, CT 06520-8035, USA
| | - Tih-Shih W Lee
- Department of Psychiatry Yale School of Medicine, New Haven, CT 06520, USA
| | - Hitten P Zaveri
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tore Eid
- Department of Laboratory Medicine, Yale School of Medicine, 330 Cedar St., PO Box 208083, New Haven, CT 06520-8035, USA.
| |
Collapse
|
30
|
Chung S, Spruston N, Koh S. Age-dependent changes in intrinsic neuronal excitability in subiculum after status epilepticus. PLoS One 2015; 10:e0119411. [PMID: 25775210 PMCID: PMC4361192 DOI: 10.1371/journal.pone.0119411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 01/30/2015] [Indexed: 12/03/2022] Open
Abstract
Kainic acid-induced status epilepticus (KA-SE) in mature rats results in the development of spontaneous recurrent seizures and a pattern of cell death resembling hippocampal sclerosis in patients with temporal lobe epilepsy. In contrast, KA-SE in young animals before postnatal day (P) 18 is less likely to cause cell death or epilepsy. To investigate whether changes in neuronal excitability occur in the subiculum after KA-SE, we examined the age-dependent effects of SE on the bursting neurons of subiculum, the major output region of the hippocampus. Patch-clamp recordings were used to monitor bursting in pyramidal neurons in the subiculum of rat hippocampal slices. Neurons were studied either one or 2-3 weeks following injection of KA or saline (control) in immature (P15) or more mature (P30) rats, which differ in their sensitivity to KA as well as the long-term sequelae of the KA-SE. A significantly greater proportion of subicular pyramidal neurons from P15 rats were strong-bursting neurons and showed increased frequency-dependent bursting compared to P30 animals. Frequency-dependent burst firing was enhanced in P30, but not in P15 rats following KA-SE. The enhancement of bursting induced by KA-SE in more mature rats suggests that the frequency-dependent limitation of repetitive burst firing, which normally occurs in the subiculum, is compromised following SE. These changes could facilitate the initiation of spontaneous recurrent seizures or their spread from the hippocampus to other parts of the brain.
Collapse
Affiliation(s)
- Sungkwon Chung
- Department of Physiology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Nelson Spruston
- Scientific Program, Janelia Research Campus, Ashburn, Virginia, United States of America
| | - Sookyong Koh
- Neurobiology Program, Stanley Manne Children’s Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
31
|
Animal models of temporal lobe epilepsy following systemic chemoconvulsant administration. J Neurosci Methods 2015; 260:45-52. [PMID: 25769270 DOI: 10.1016/j.jneumeth.2015.03.009] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 01/23/2023]
Abstract
In order to understand the pathophysiology of temporal lobe epilepsy (TLE), and thus to develop new pharmacological treatments, in vivo animal models that present features similar to those seen in TLE patients have been developed during the last four decades. Some of these models are based on the systemic administration of chemoconvulsants to induce an initial precipitating injury (status epilepticus) that is followed by the appearance of recurrent seizures originating from limbic structures. In this paper we will review two chemically-induced TLE models, namely the kainic acid and pilocarpine models, which have been widely employed in basic epilepsy research. Specifically, we will take into consideration their behavioral, electroencephalographic and neuropathologic features. We will also evaluate the response of these models to anti-epileptic drugs and the impact they might have in developing new treatments for TLE.
Collapse
|
32
|
Lévesque M, Behr C, Avoli M. The anti-ictogenic effects of levetiracetam are mirrored by interictal spiking and high-frequency oscillation changes in a model of temporal lobe epilepsy. Seizure 2014; 25:18-25. [PMID: 25645630 DOI: 10.1016/j.seizure.2014.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/18/2014] [Accepted: 11/20/2014] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Mesial temporal lobe epilepsy (MTLE) is the most prevalent type of partial epileptic disorders. In this study, we have analyzed the impact of levetiracetam (LEV) in the pilocarpine model of MTLE. METHODS Sprague-Dawley rats (n=19) were injected with pilocarpine (380 mg/kg, i.p.) to induce a status epilepticus. Twelve animals were used as controls and seven were treated with LEV. They were implanted with bipolar electrodes in the CA3 subfield of the hippocampus, entorhinal cortex (EC), dentate gyrus (DG) and subiculum and EEG-video monitored continuously from day 4 to day 14 after SE. RESULTS Only 29% of LEV-treated animals had seizures compared to all controls following a latent period that was similar in duration. Seizure rates were lower in LEV-treated animals. In LEV-treated animals without seizures, lower interictal spike rates were found in all regions compared to controls. Analysis of interictal high-frequency oscillations (HFO s) revealed that LEV-treated animals without seizures had lower rates of interictal spikes with ripples (80-200 Hz) in CA3, EC and subiculum (p<0.01), whereas rates of interictal spikes with fast ripples (250-500 Hz) were significantly lower in CA3 and subiculum, compared to controls. CONCLUSION Our findings indicate that the anti-ictogenic properties of LEV are mirrored by decreases of interictal spike rate in temporal lobe regions, and are accompanied by subregion-specific decreases of HFO occurrence in CA3 and subiculum. Overall, this evidence suggest that LEV may inhibit neural network activity in regions that are known to play important roles in MTLE.
Collapse
Affiliation(s)
- Maxime Lévesque
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, Montréal, Qc H3A 2B4, Canada
| | - Charles Behr
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, Montréal, Qc H3A 2B4, Canada
| | - Massimo Avoli
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, Montréal, Qc H3A 2B4, Canada.
| |
Collapse
|
33
|
Galanopoulou AS, Moshé SL. Does epilepsy cause a reversion to immature function? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:195-209. [PMID: 25012378 DOI: 10.1007/978-94-017-8914-1_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Seizures have variable effects on brain. Numerous studies have examined the consequences of seizures, in light of the way that these may alter the susceptibility of the brain to seizures, promote epileptogenesis, or functionally alter brain leading to seizure-related comorbidities. In many -but not all- situations, seizures shift brain function towards a more immature state, promoting the birth of newborn neurons, altering the dendritic structure and neuronal connectivity, or changing neurotransmitter signaling towards more immature patterns. These effects depend upon many factors, including the seizure type, age of seizure occurrence, sex, and brain region studied. Here we discuss some of these findings proposing that these seizure-induced immature features do not simply represent rejuvenation of the brain but rather a de-synchronization of the homeostatic mechanisms that were in place to maintain normal physiology, which may contribute to epileptogenesis or the cognitive comorbidities.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, The Laboratory of Developmental Epilepsy, Comprehensive Einstein/Montefiore Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 306, Bronx, NY, 10461, USA,
| | | |
Collapse
|
34
|
Curia G, Lucchi C, Vinet J, Gualtieri F, Marinelli C, Torsello A, Costantino L, Biagini G. Pathophysiogenesis of mesial temporal lobe epilepsy: is prevention of damage antiepileptogenic? Curr Med Chem 2014; 21:663-88. [PMID: 24251566 PMCID: PMC4101766 DOI: 10.2174/0929867320666131119152201] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/04/2013] [Accepted: 08/29/2013] [Indexed: 12/26/2022]
Abstract
Temporal lobe epilepsy (TLE) is frequently associated with hippocampal sclerosis, possibly caused by a primary brain injury that occurred a long time before the appearance of neurological symptoms. This type of epilepsy is characterized by refractoriness to drug treatment, so to require surgical resection of mesial temporal regions involved in seizure onset. Even this last therapeutic approach may fail in giving relief to patients. Although prevention of hippocampal damage and epileptogenesis after a primary event could be a key innovative approach to TLE, the lack of clear data on the pathophysiological mechanisms leading to TLE does not allow any rational therapy. Here we address the current knowledge on mechanisms supposed to be involved in epileptogenesis, as well as on the possible innovative treatments that may lead to a preventive approach. Besides loss of principal neurons and of specific interneurons, network rearrangement caused by axonal sprouting and neurogenesis are well known phenomena that are integrated by changes in receptor and channel functioning and modifications in other cellular components. In particular, a growing body of evidence from the study of animal models suggests that disruption of vascular and astrocytic components of the blood-brain barrier takes place in injured brain regions such as the hippocampus and piriform cortex. These events may be counteracted by drugs able to prevent damage to the vascular component, as in the case of the growth hormone secretagogue ghrelin and its analogues. A thoroughly investigation on these new pharmacological tools may lead to design effective preventive therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - G Biagini
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Laboratorio di Epilettologia Sperimentale, Universita di Modena e Reggio Emilia, Via Campi, 287, 41125 Modena, Italy.
| |
Collapse
|
35
|
Drexel M, Kirchmair E, Sperk G. Changes in the expression of GABAA receptor subunit mRNAs in parahippocampal areas after kainic acid induced seizures. Front Neural Circuits 2013; 7:142. [PMID: 24065890 PMCID: PMC3776158 DOI: 10.3389/fncir.2013.00142] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/24/2013] [Indexed: 11/30/2022] Open
Abstract
The parahippocampal areas including the subiculum, pre- and parasubiculum, and notably the entorhinal cortex (EC) are intimately involved in the generation of limbic seizures in temporal lobe epilepsy. We investigated changes in the expression of 10 major GABAA receptor subunit mRNAs in subfields of the ventral hippocampus, ventral subiculum, EC, and perirhinal cortex (PRC) at different intervals (1, 8, 30, and 90 days) after kainic acid (KA)-induced status epilepticus priming epileptogenesis in the rat. The most pronounced and ubiquitous changes were a transient (24 h after KA only) down-regulation of γ2 mRNA and lasting decreases in subunit α5, β3, and δ mRNAs that were prominent in all hippocampal and parahippocampal areas. In the subiculum similarly as in sectors CA1 and CA3, levels of subunit α1, α2, α4, and γ2 mRNAs decreased transiently (1 day after KA-induced status epilepticus). They were followed by increased expression of subunit α1 and α3 mRNAs in the dentate gyrus (DG) and sectors CA1 and CA3, and subunit α1 also in the EC layer II (30 and 90 days after KA). We also observed sustained overexpression of subunits α4 and γ2 in the subiculum and in the Ammon’s horn. Subunit γ2 mRNA was also increased in sector CA1 at the late intervals after KA. Taken together, our results suggest distinct regulation of mRNA expression for individual GABAA receptor subunits. Especially striking was the wide-spread down-regulation of the often peri- or extrasynaptically located subunits α5 and δ. These subunits are often associated with tonic inhibition. Their decrease could be related to decreased tonic inhibition or may merely reflect compensatory changes. In contrast, expression of subunit α4 that may also mediate tonic inhibition when associated with the δ-subunit was significantly upregulated in the DG and in the proximal subiculum at late intervals. Thus, concomitant up-regulation of subunit γ2, α1 and α4 mRNAs (and loss in δ-subunits) ultimately indicates significant rearrangement of GABAA receptor composition after KA-induced seizures.
Collapse
Affiliation(s)
- Meinrad Drexel
- Department of Pharmacology, Innsbruck Medical University Innsbruck, Austria
| | | | | |
Collapse
|
36
|
Biagini G, D'Antuono M, Benini R, de Guzman P, Longo D, Avoli M. Perirhinal cortex and temporal lobe epilepsy. Front Cell Neurosci 2013; 7:130. [PMID: 24009554 PMCID: PMC3756799 DOI: 10.3389/fncel.2013.00130] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/01/2013] [Indexed: 12/30/2022] Open
Abstract
The perirhinal cortex—which is interconnected with several limbic structures and is intimately involved in learning and memory—plays major roles in pathological processes such as the kindling phenomenon of epileptogenesis and the spread of limbic seizures. Both features may be relevant to the pathophysiology of mesial temporal lobe epilepsy that represents the most refractory adult form of epilepsy with up to 30% of patients not achieving adequate seizure control. Compared to other limbic structures such as the hippocampus or the entorhinal cortex, the perirhinal area remains understudied and, in particular, detailed information on its dysfunctional characteristics remains scarce; this lack of information may be due to the fact that the perirhinal cortex is not grossly damaged in mesial temporal lobe epilepsy and in models mimicking this epileptic disorder. However, we have recently identified in pilocarpine-treated epileptic rats the presence of selective losses of interneuron subtypes along with increased synaptic excitability. In this review we: (i) highlight the fundamental electrophysiological properties of perirhinal cortex neurons; (ii) briefly stress the mechanisms underlying epileptiform synchronization in perirhinal cortex networks following epileptogenic pharmacological manipulations; and (iii) focus on the changes in neuronal excitability and cytoarchitecture of the perirhinal cortex occurring in the pilocarpine model of mesial temporal lobe epilepsy. Overall, these data indicate that perirhinal cortex networks are hyperexcitable in an animal model of temporal lobe epilepsy, and that this condition is associated with a selective cellular damage that is characterized by an age-dependent sensitivity of interneurons to precipitating injuries, such as status epilepticus.
Collapse
Affiliation(s)
- Giuseppe Biagini
- Laboratory of Experimental Epileptology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia Modena, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Huang L, van Luijtelaar G. The effects of responsive and scheduled subicular high frequency stimulation in the intra-hippocampal kainic acid seizure model. Epilepsy Res 2013; 106:326-37. [PMID: 23899954 DOI: 10.1016/j.eplepsyres.2013.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 05/22/2013] [Accepted: 06/25/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Responsive stimulation is a promising and newly emerging treatment for refractory temporal lobe epilepsy in which current is delivered to target areas following seizure occurrence. OBJECTIVE We compared responsive and scheduled subicular high frequency stimulation (HFS) with a sham control group on acute seizures and seizure sensitivity two weeks later. We also investigated the role of status epilepticus (SE) on efficacy of both types of stimulation. METHOD Adult Wistar rats received kainic acid (KA) injections intrahippocampally until they reached Stage V (Racine scale) on Day 1. Responsive, scheduled or sham HFS (125 Hz, 100 μs) was delivered in three groups while EEG was recorded. All rats received KA injections again on Day 15 to measure the excitability of animals to KA, again with EEG monitoring. RESULTS All rats reached Stage V and 60% reached SE on Day 1. Focal seizures were suppressed in both stimulated groups (the scheduled group was slightly more effective) on both days in only non-SE rats. Similar stimulation effects were found on generalized seizures but mainly on Day 15. CONCLUSION Both types of subicular HFS suppressed focal and generalized seizures, albeit differently. Scheduled stimulation seemed a bit more effective, and the amount of stimulation might be a factor that influences the differences between the stimulated groups. Beneficial effects of HFS were restricted to non-SE rats and HFS did not suppress or even worsen seizures in SE rats.
Collapse
Affiliation(s)
- L Huang
- Department of Biological Psychology, Donders Center for Cognition, Donders Institute for Brain Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands.
| | | |
Collapse
|
38
|
Sah N, Sikdar SK. Transition in subicular burst firing neurons from epileptiform activity to suppressed state by feedforward inhibition. Eur J Neurosci 2013; 38:2542-56. [DOI: 10.1111/ejn.12262] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 04/21/2013] [Accepted: 04/23/2013] [Indexed: 01/03/2023]
Affiliation(s)
- Nirnath Sah
- Molecular Biophysics Unit; Indian Institute of Science; Bangalore; India
| | - Sujit K. Sikdar
- Molecular Biophysics Unit; Indian Institute of Science; Bangalore; India
| |
Collapse
|
39
|
Curia G, Gualtieri F, Bartolomeo R, Vezzali R, Biagini G. Resilience to audiogenic seizures is associated with p-ERK1/2 dephosphorylation in the subiculum of Fmr1 knockout mice. Front Cell Neurosci 2013; 7:46. [PMID: 23630463 PMCID: PMC3635025 DOI: 10.3389/fncel.2013.00046] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 04/03/2013] [Indexed: 12/30/2022] Open
Abstract
Young, but not adult, fragile X mental retardation gene (Fmr1) knockout (KO) mice display audiogenic seizures (AGS) that can be prevented by inhibiting extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation. In order to identify the cerebral regions involved in these phenomena, we characterized the response to AGS in Fmr1 KO mice and wild type (WT) controls at postnatal day (P) 45 and P90. To characterize the diverse response to AGS in various cerebral regions, we evaluated the activity markers FosB/ΔFosB and phosphorylated ERK1/2 (p-ERK1/2). Wild running (100% of tested mice) followed by clonic/tonic seizures (30%) were observed in P45 Fmr1 KO mice, but not in WT mice. In P90 Fmr1 KO mice, wild running was only present in 25% of tested animals. Basal FosB/ΔFosB immunoreactivity was higher (P < 0.01 vs. WT) in the CA1 and subiculum of P45 Fmr1 KO mice. Following the AGS test, FosB/ΔFosB expression consistently increased in most of the analyzed regions in both groups at P45, but not at P90. Interestingly, FosB/ΔFosB immunoreactivity was significantly higher in P45 Fmr1 KO mice in the medial geniculate body (P < 0.05 vs. WT) and CA3 (P < 0.01). Neurons presenting with immunopositivity to p-ERK1/2 were more abundant in the subiculum of Fmr1 KO mice in control condition (P < 0.05 vs. WT, in both age groups). In this region, p-ERK1/2-immunopositive cells significantly decreased (–75%, P < 0.01) in P90 Fmr1 KO mice exposed to the AGS test, but no changes were found in P45 mice or in other brain regions. In both age groups of WT mice, p-ERK1/2-immunopositive cells increased in the subiculum after exposure to the acoustic test. Our findings illustrate that FosB/ΔFosB markers are overexpressed in the medial geniculate body and CA3 in Fmr1 KO mice experiencing AGS, and that p-ERK1/2 is markedly decreased in the subiculum of Fmr1 KO mice resistant to AGS induction. These findings suggest that resilience to AGS is associated with dephosphorylation of p-ERK1/2 in the subiculum of mature Fmr1 KO mice.
Collapse
Affiliation(s)
- Giulia Curia
- Laboratory of Experimental Epileptology, Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia Modena, Italy
| | | | | | | | | |
Collapse
|
40
|
Cell type-specific properties of subicular GABAergic currents shape hippocampal output firing mode. PLoS One 2012; 7:e50241. [PMID: 23251362 PMCID: PMC3519474 DOI: 10.1371/journal.pone.0050241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 10/22/2012] [Indexed: 11/19/2022] Open
Abstract
GABAergic function of the subiculum is central to the regulation of hippocampal output activity. Subicular neuronal networks are indeed under potent control by local inhibition. However, information about the properties of GABAergic currents generated by neurons of this parahippocampal area in normal tissue is still missing. Here, we describe GABAA receptor (GABAAR)-mediated phasic and tonic currents generated by principal cells (PCs) and interneurons (INs) of the rat subiculum. We show that in spite of similar synaptic current densities, INs generate spontaneous IPSCs (sIPSCs) that occur less frequently and exhibit smaller charge transfer, thus receiving less synaptic total current than PCs. Further distinction of PCs between intrinsically bursting (IB) and regular-spiking (RS) neurons suggested that sIPSCs generated by the two PC sub-types are likely to be similar. PCs and INs are also controlled by a similar tonic inhibition. However, whereas a comparable tonic current density is found in RS cells and INs, IB neurons are constrained by a greater inhibitory tone. Finally, pharmacological blockade of GABAAR did not promote functional switch of RS neurons to IB mode, but influenced the bursting propensity of IB cells and released fast spiking activity in INs. Our findings reveal differences in GABAergic currents between PCs and INs as well as within PC sub-types. We propose that GABAergic inhibition may shape hippocampal output activity by providing cell type-specific fine-tuning of subicular excitatory and inhibitory drives.
Collapse
|
41
|
Deeb TZ, Maguire J, Moss SJ. Possible alterations in GABAA receptor signaling that underlie benzodiazepine-resistant seizures. Epilepsia 2012; 53 Suppl 9:79-88. [PMID: 23216581 PMCID: PMC4402207 DOI: 10.1111/epi.12037] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Benzodiazepines have been used for decades as first-line treatment for status epilepticus (SE). For reasons that are not fully understood, the efficacy of benzodiazepines decreases with increasing duration of seizure activity. This often forces clinicians to resort to more drastic second- and third-line treatments that are not always successful. The antiseizure properties of benzodiazepines are mediated by γ-aminobutyric acid type A (GABA(A) ) receptors. Decades of research have focused on the failure of GABAergic inhibition after seizure onset as the likely cause of the development benzodiazepine resistance during SE. However, the details of the deficits in GABA(A) signaling are still largely unknown. Therefore, it is necessary to improve our understanding of the mechanisms of benzodiazepine resistance so that more effective strategies can be formulated. In this review we discuss evidence supporting the role of altered GABA(A) receptor function as the major underlying cause of benzodiazepine-resistant SE in both humans and animal models. We specifically address the prevailing hypothesis, which is based on changes in the number and subtypes of GABA(A) receptors, as well as the potential influence of perturbed chloride homeostasis in the mature brain.
Collapse
Affiliation(s)
- Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
42
|
Huang L, Luijtelaar G. The effects of acute responsive high frequency stimulation of the subiculum on the intra-hippocampal kainic acid seizure model in rats. Brain Behav 2012; 2:532-40. [PMID: 23139899 PMCID: PMC3489806 DOI: 10.1002/brb3.70] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 02/23/2012] [Accepted: 05/11/2012] [Indexed: 11/05/2022] Open
Abstract
The effects of acute responsive high frequency stimulation (HFS) to the subiculum on seizures and interictal spikes were investigated in a semi-acute kainic acid (KA) induced seizure model in rats. Wistar rats (n = 15) were implanted with an electrode-cannula complex in the CA3 area, stimulation and recording electrodes in the subiculum and another recording electrode at the contralateral motor cortex. Two weeks later rats were injected repeatedly with KA (0.05 μg/0.1 μL) for 3 days with an interval of 48 h. HFS (125 Hz, 100 μsec) was delivered to the subiculum at a predetermined intensity range (100-500 μA) in the HFS group (n = 7) when seizures were visually detected, while no stimulation was delivered in the sham control group (n = 8). Various severities of seizures were obtained (Stage I-V) and all rats of both groups reached Stage V (Racine's scale) on Day 1. The HFS group had less focal seizures and a longer inter-focal seizure interval on Day 1. Interictal spike rate was also lower in the HFS group and decreased with injection days. Significant day effects were found for the latency, number of focal seizures, and duration of focal seizures and generalized seizures while differences between groups were no longer present. Responsive HFS did not disrupt ongoing seizures. However, focal seizures and interictal spikes were suppressed by HFS. Such anticonvulsant effects of acute subicular stimulation indicate that the subiculum is involved in seizure generation. The reduction of seizure sensitivity over the injection day reflects an intrinsic anticonvulsant mechanism.
Collapse
Affiliation(s)
- L Huang
- Department of Biological Psychology, Donders Center for Cognition, Donders Institute for Brain Cognition and Behaviour, Radboud University Nijmegen Nijmegen, The Netherlands
| | | |
Collapse
|
43
|
Drexel M, Preidt AP, Sperk G. Sequel of spontaneous seizures after kainic acid-induced status epilepticus and associated neuropathological changes in the subiculum and entorhinal cortex. Neuropharmacology 2012; 63:806-17. [PMID: 22722023 PMCID: PMC3409872 DOI: 10.1016/j.neuropharm.2012.06.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 05/15/2012] [Accepted: 06/08/2012] [Indexed: 12/31/2022]
Abstract
Injection of the seaweed toxin kainic acid (KA) in rats induces a severe status epilepticus initiating complex neuropathological changes in limbic brain areas and subsequently spontaneous recurrent seizures. Although neuropathological changes have been intensively investigated in the hippocampus proper and the dentate gyrus in various seizure models, much less is known about changes in parahippocampal areas. We now established telemetric EEG recordings combined with continuous video monitoring to characterize the development of spontaneous seizures after KA-induced status epilepticus, and investigated associated neurodegenerative changes, astrocyte and microglia proliferation in the subiculum and other parahippocampal brain areas. The onset of spontaneous seizures was heterogeneous, with an average latency of 15 ± 1.4 days (range 3–36 days) to the initial status epilepticus. The frequency of late spontaneous seizures was higher in rats in which the initial status epilepticus was recurrent after its interruption with diazepam compared to rats in which this treatment was more efficient. Seizure-induced neuropathological changes were assessed in the subiculum by losses in NeuN-positive neurons and by Fluoro-Jade C staining of degenerating neurons. Neuronal loss was already prominent 24 h after KA injection and only modestly progressed at the later intervals. It was most severe in the proximal subiculum and in layer III of the medial entorhinal cortex and distinct Fluoro-Jade C labeling was observed there in 75% of rats even after 3 months. Glutamatergic neurons, labeled by in situ hybridization for the vesicular glutamate transporter 1 followed a similar pattern of cell losses, except for the medial entorhinal cortex and the proximal subiculum that appeared more vulnerable. Glutamate decarboxylase65 (GAD65) mRNA expressing neurons were generally less vulnerable than glutamate neurons. Reactive astrocytes and microglia were present after 24 h, however, became prominent only after 8 days and remained high after 30 days. In the proximal subiculum, parasubiculum and entorhinal cortex the number of microglia cells was highest after 30 days. Although numbers of reactive astrocytes and microglia were reduced again after 3 months, they were still present in most rats. The time course of astrocyte and microglia proliferation parallels that of epileptogenesis.
Collapse
Affiliation(s)
- Meinrad Drexel
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr-Str. 1a, 6020 Innsbruck, Austria.
| | | | | |
Collapse
|
44
|
Pyramidal cells accumulate chloride at seizure onset. Neurobiol Dis 2012; 47:358-66. [PMID: 22677032 DOI: 10.1016/j.nbd.2012.05.016] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 05/21/2012] [Accepted: 05/24/2012] [Indexed: 01/09/2023] Open
Abstract
Seizures are thought to originate from a failure of inhibition to quell hyperactive neural circuits, but the nature of this failure remains unknown. Here we combine high-speed two-photon imaging with electrophysiological recordings to directly evaluate the interaction between populations of interneurons and principal cells during the onset of seizure-like activity in mouse hippocampal slices. Both calcium imaging and dual patch clamp recordings reveal that in vitro seizure-like events (SLEs) are preceded by pre-ictal bursts of activity in which interneurons predominate. Corresponding changes in intracellular chloride concentration were observed in pyramidal cells using the chloride indicator Clomeleon. These changes were measurable at SLE onset and became very large during the SLE. Pharmacological manipulation of GABAergic transmission, either by blocking GABA(A) receptors or by hyperpolarizing the GABA(A) reversal potential, converted SLEs to short interictal-like bursts. Together, our results support a model in which pre-ictal GABA(A) receptor-mediated chloride influx shifts E(GABA) to produce a positive feedback loop that contributes to the initiation of seizure activity.
Collapse
|
45
|
Wide therapeutic time-window of low-frequency stimulation at the subiculum for temporal lobe epilepsy treatment in rats. Neurobiol Dis 2012; 48:20-6. [PMID: 22659307 DOI: 10.1016/j.nbd.2012.05.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 05/07/2012] [Accepted: 05/24/2012] [Indexed: 11/22/2022] Open
Abstract
Low-frequency stimulation (LFS) has been considered as an option for the treatment of intractable epilepsy. However, previous data showed that LFS of certain brain regions only exerts its effect within a very narrow therapeutic time window, which lasts from seconds to tens of seconds, thus restricting its clinical application. The present study was designed to determine whether there exists a target with a wider therapeutic window for LFS treatment. Therefore, evoked seizures in the rat were induced by amygdala kindling and spontaneous seizures were induced by pilocarpine. The effects of different modes of LFS at the subiculum on the progression and severity of evoked seizures and the frequency of spontaneous seizure were evaluated. We found that (i) LFS at 1Hz delivered to the subiculum before and immediately after the kindling stimulations, or after the cessation of afterdischarge (afterdischarge duration, ADD) decreased the seizure stages and shortened the ADD both in seizure acquisition and expression in amygdaloid-kindled seizures. In addition, even LFS delivered after duration of double the ADD prolonged the kindling progression. (ii) LFS delivered at 1Hz, but not 0.5, 3 or 130Hz, immediately after the cessation of kindling stimulations retarded the progression of kindling seizures. (iii) Pilocarpine-induced spontaneous seizures were completely inhibited by 1Hz LFS. Thus, these results demonstrated that LFS of the subiculum has a wide therapeutic time-window for temporal lobe epilepsy treatment in rats, suggesting that the subiculum may be a promising and suitable target for clinical application.
Collapse
|
46
|
Somatostatin and neuropeptide Y neurons undergo different plasticity in parahippocampal regions in kainic acid-induced epilepsy. J Neuropathol Exp Neurol 2012; 71:312-29. [PMID: 22437342 DOI: 10.1097/nen.0b013e31824d9882] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Parahippocampal brain areas including the subiculum, presubiculum and parasubiculum, and entorhinal cortex give rise to major input and output neurons of the hippocampus and exert increased excitability in animal models and human temporal lobe epilepsy. Using immunohistochemistry and in situ hybridization for somatostatin and neuropeptide Y, we investigated plastic morphologic and neurochemical changes in parahippocampal neurons in the kainic acid (KA) model of temporal lobe epilepsy. Although constitutively contained in similar subclasses of γ-aminobutyric acid (GABA)-ergic neurons, both neuropeptide systems undergo distinctly different changes in their expression. Somatostatin messenger RNA (mRNA) is rapidly but transiently expressed de novo in pyramidal neurons of the subiculum and entorhinal cortex 24 hours after KA. Surviving somatostatin interneurons display increased mRNA levels at late intervals (3 months) after KA and increased labeling of their terminals in the outer molecular layer of the subiculum; the labeling correlates with the number of spontaneous seizures, suggesting that the seizures may trigger somatostatin expression. In contrast, neuropeptide Y mRNA is consistently expressed in principal neurons of the proximal subiculum and the lateral entorhinal cortex and labeling for the peptide persistently increased in virtually all major excitatory pathways of the hippocampal formation. The pronounced plastic changes differentially involving both neuropeptide systems indicate marked rearrangement of parahippocampal areas, presumably aiming at endogenous seizure protection. Their receptors may be targets for anticonvulsive drug therapy.
Collapse
|
47
|
Avoli M, de Curtis M. GABAergic synchronization in the limbic system and its role in the generation of epileptiform activity. Prog Neurobiol 2011; 95:104-32. [PMID: 21802488 PMCID: PMC4878907 DOI: 10.1016/j.pneurobio.2011.07.003] [Citation(s) in RCA: 190] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 07/14/2011] [Accepted: 07/15/2011] [Indexed: 11/30/2022]
Abstract
GABA is the main inhibitory neurotransmitter in the adult forebrain, where it activates ionotropic type A and metabotropic type B receptors. Early studies have shown that GABA(A) receptor-mediated inhibition controls neuronal excitability and thus the occurrence of seizures. However, more complex, and at times unexpected, mechanisms of GABAergic signaling have been identified during epileptiform discharges over the last few years. Here, we will review experimental data that point at the paradoxical role played by GABA(A) receptor-mediated mechanisms in synchronizing neuronal networks, and in particular those of limbic structures such as the hippocampus, the entorhinal and perirhinal cortices, or the amygdala. After having summarized the fundamental characteristics of GABA(A) receptor-mediated mechanisms, we will analyze their role in the generation of network oscillations and their contribution to epileptiform synchronization. Whether and how GABA(A) receptors influence the interaction between limbic networks leading to ictogenesis will be also reviewed. Finally, we will consider the role of altered inhibition in the human epileptic brain along with the ability of GABA(A) receptor-mediated conductances to generate synchronous depolarizing events that may lead to ictogenesis in human epileptic disorders as well.
Collapse
Affiliation(s)
- Massimo Avoli
- Montreal Neurological Institute and Departments of Neurology & Neurosurgery, and of Physiology, McGill University, Montreal H3A 2B4 Quebec, Canada.
| | | |
Collapse
|
48
|
Lévesque M, Bortel A, Gotman J, Avoli M. High-frequency (80-500 Hz) oscillations and epileptogenesis in temporal lobe epilepsy. Neurobiol Dis 2011; 42:231-41. [PMID: 21238589 PMCID: PMC4873283 DOI: 10.1016/j.nbd.2011.01.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 11/17/2010] [Accepted: 01/02/2011] [Indexed: 11/25/2022] Open
Abstract
High-frequency oscillations (HFOs), termed ripples (80-200 Hz) and fast ripples (250-600 Hz), are recorded in the EEG of epileptic patients and in animal epilepsy models; HFOs are thought to reflect pathological activity and seizure onset zones. Here, we analyzed the temporal and spatial evolution of interictal spikes with and without HFOs in the rat pilocarpine model of temporal lobe epilepsy. Depth electrode recordings from dentate gyrus (DG), CA3 region, subiculum and entorhinal cortex (EC), were obtained from rats between the 4th and 15th day after a status epilepticus (SE) induced by i.p. injection of pilocarpine. The first seizure occurred 6.1 ± 2.5 days after SE (n = 7 rats). Five of 7 animals exhibited interictal spikes that co-occurred with fast ripples accounting for 4.9 ± 4.6% of all analyzed interictal spikes (n = 12,886) while all rats showed interictal spikes co-occurring with ripples, accounting for 14.3 ± 3.4% of all events. Increased rates of interictal spikes without HFOs in the EC predicted upcoming seizures on the following day, while rates of interictal spikes with fast ripples in CA3 reflected periods of high seizure occurrence. Finally, interictal spikes co-occurring with ripples did not show any specific relation to seizure occurrence. Our findings identify different temporal and spatial developmental patterns for the rates of interictal spikes with or without HFOs in relation with seizure occurrence. These distinct categories of interictal spikes point at dynamic processes that should bring neuronal networks close to seizure generation.
Collapse
Affiliation(s)
- Maxime Lévesque
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montreal, Qc, Canada H3A 2B4
| | - Aleksandra Bortel
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montreal, Qc, Canada H3A 2B4
| | - Jean Gotman
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montreal, Qc, Canada H3A 2B4
| | - Massimo Avoli
- Montreal Neurological Institute and Department of Neurology & Neurosurgery, McGill University, 3801 University Street, Montreal, Qc, Canada H3A 2B4
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Viale del Castro Laurenziano 9, 00185 Roma, Italy
| |
Collapse
|
49
|
Drexel M, Preidt AP, Kirchmair E, Sperk G. Parvalbumin interneurons and calretinin fibers arising from the thalamic nucleus reuniens degenerate in the subiculum after kainic acid-induced seizures. Neuroscience 2011; 189:316-29. [PMID: 21616128 PMCID: PMC3152681 DOI: 10.1016/j.neuroscience.2011.05.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 05/03/2011] [Accepted: 05/11/2011] [Indexed: 12/20/2022]
Abstract
The subiculum is the major output area of the hippocampus. It is closely interconnected with the entorhinal cortex and other parahippocampal areas. In animal models of temporal lobe epilepsy (TLE) and in TLE patients it exerts increased network excitability and may crucially contribute to the propagation of limbic seizures. Using immunohistochemistry and in situ-hybridization we now investigated neuropathological changes affecting parvalbumin and calretinin containing neurons in the subiculum and other parahippocampal areas after kainic acid-induced status epilepticus. We observed prominent losses in parvalbumin containing interneurons in the subiculum and entorhinal cortex, and in the principal cell layers of the pre- and parasubiculum. Degeneration of parvalbumin-positive neurons was associated with significant precipitation of parvalbumin-immunoreactive debris 24 h after kainic acid injection. In the subiculum the superficial portion of the pyramidal cell layer was more severely affected than its deep part. In the entorhinal cortex, the deep layers were more severely affected than the superficial ones. The decrease in number of parvalbumin-positive neurons in the subiculum and entorhinal cortex correlated with the number of spontaneous seizures subsequently experienced by the rats. The loss of parvalbumin neurons thus may contribute to the development of spontaneous seizures. On the other hand, surviving parvalbumin neurons revealed markedly increased expression of parvalbumin mRNA notably in the pyramidal cell layer of the subiculum and in all layers of the entorhinal cortex. This indicates increased activity of these neurons aiming to compensate for the partial loss of this functionally important neuron population. Furthermore, calretinin-positive fibers terminating in the molecular layer of the subiculum, in sector CA1 of the hippocampus proper and in the entorhinal cortex degenerated together with their presumed perikarya in the thalamic nucleus reuniens. In addition, a significant loss of calretinin containing interneurons was observed in the subiculum. Notably, the loss in parvalbumin positive neurons in the subiculum equaled that in human TLE. It may result in marked impairment of feed-forward inhibition of the temporo-ammonic pathway and may significantly contribute to epileptogenesis. Similarly, the loss of calretinin-positive fiber tracts originating from the nucleus reuniens thalami significantly contributes to the rearrangement of neuronal circuitries in the subiculum and entorhinal cortex during epileptogenesis.
Collapse
Affiliation(s)
- M Drexel
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr-Str. 1a, 6020 Innsbruck, Austria.
| | | | | | | |
Collapse
|
50
|
A knock-in reporter mouse model for Batten disease reveals predominant expression of Cln3 in visual, limbic and subcortical motor structures. Neurobiol Dis 2010; 41:237-48. [PMID: 20875858 DOI: 10.1016/j.nbd.2010.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/30/2010] [Accepted: 09/19/2010] [Indexed: 11/23/2022] Open
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL) or Batten disease is an autosomal recessive neurodegenerative disorder of children caused by mutation in CLN3. JNCL is characterized by progressive visual impairment, cognitive and motor deficits, seizures and premature death. Information about the localization of CLN3 expressing neurons in the nervous system is limited, especially during development. The present study has systematically mapped the spatial and temporal localization of CLN3 reporter neurons in the entire nervous system including retina, using a knock-in reporter mouse model. CLN3 reporter is expressed predominantly in post-migratory neurons in visual and limbic cortices, anterior and intralaminar thalamic nuclei, amygdala, cerebellum, red nucleus, reticular formation, vestibular nuclei and retina. CLN3 reporter in the nervous system is mainly expressed during the first postnatal month except in the dentate gyrus, parasolitary nucleus and retina, where it is still strongly expressed in adulthood. The predominant distribution of CLN3 reporter neurons in visual, limbic and subcortical motor structures correlates well with the clinical symptoms of JNCL. These findings have also revealed potential target brain regions and time periods for future investigations of the disease mechanisms and therapeutic intervention.
Collapse
|