1
|
Khan T, Hussain AI, Casilli TP, Frayser L, Cho M, Williams G, McFall D, Forcelli PA. Prophylactic senolytic treatment in aged mice reduces seizure severity and improves survival from Status Epilepticus. Aging Cell 2024; 23:e14239. [PMID: 39031751 PMCID: PMC11488304 DOI: 10.1111/acel.14239] [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: 01/19/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 07/22/2024] Open
Abstract
Increased vulnerability to seizures in aging has been well documented both clinically and in various models of aging in epilepsy. Seizures can exacerbate cognitive decline that is already prominent in aging. Senescent cells are thought to contribute to cognitive impairment in aging and clearing senescent cells with senolytic drugs improves cognitive function in animal models. It remains unclear whether senescent cells render the aged brain vulnerable to seizures. Here, we demonstrate that prophylactic senolytic treatment with Dasatinib and Quercetin (D&Q) reduced both seizure severity and mortality in aged C57BL/6J mice. We subjected the D&Q and VEH-treated aged mice to spatial memory testing before and after an acute seizure insult, Status Epilepticus [SE], which leads to epilepsy development. We found that senolytic therapy improved spatial memory before injury, however, spatial memory was not rescued after SE. Senescence-related proteins p16 and senescence-associated β-galactosidase were reduced in D&Q-treated aged mice. Our findings indicate that senescent cells increase seizure susceptibility in aging. Thus, prophylactically targeting senescent cells may prevent age-related seizure vulnerability.
Collapse
Affiliation(s)
- Tahiyana Khan
- Interdisciplinary Program in NeuroscienceGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Abbas I. Hussain
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Timothy P. Casilli
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Logan Frayser
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Michelle Cho
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Gabrielle Williams
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - David McFall
- Interdisciplinary Program in NeuroscienceGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Patrick A. Forcelli
- Department of Pharmacology and PhysiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
2
|
Yu X, Yang H, Lv H, Lu H, Zhao H, Xu Z. Age-Dependent Phenomena of 6-Hz Corneal Kindling Model in Mice. Mol Neurobiol 2024; 61:5601-5613. [PMID: 38214837 DOI: 10.1007/s12035-024-03934-x] [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: 09/21/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Although numerous studies have acknowledged disparities in epilepsy-related disease processes between young and aged animals, little is known about how epilepsy changes from young adulthood to middle age. This study investigates the impact of aging on 6-Hz corneal kindling in young-adult mice and middle-aged mice. We found that the kindling acquisition of the 6-Hz corneal kindling model was delayed in middle-aged mice when compared to young-adult mice. While the seizure stage and incidence of generalized seizures (GS) were similar between the two age groups, the duration of GS in the kindled middle-aged mice was shorter than that in the kindled young-adult mice. Besides, all kindled mice, regardless of age, were resistant to phenytoin sodium (PHT), valproate sodium (VPA), and lamotrigine (LGT), whereas middle-aged mice exhibited higher levetiracetam (LEV) resistance compared to young-adult mice. Both age groups of kindled mice displayed hyperactivity and impaired memory, which are common behavioral characteristics associated with epilepsy. Furthermore, middle-aged mice displayed more pronounced astrogliosis in the hippocampus. Additionally, the expression of Brain-Derived Neurotrophic Factor (BDNF) was lower in middle-aged mice than in young-adult mice prior to kindling. These data demonstrate that both the acquisition and expression of 6-Hz corneal kindling are attenuated in middle-aged mice, while hippocampal astrogliosis and pharmacological resistance are more pronounced in this age group. These results underscore the importance of considering age-related factors when utilizing the 6-Hz corneal kindling model in mice of varying age groups.
Collapse
Affiliation(s)
- Xiu Yu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, No.548 Binwen Road, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Han Yang
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, No.548 Binwen Road, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - HongJie Lv
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, No.548 Binwen Road, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haimei Lu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, No.548 Binwen Road, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huawei Zhao
- Department of Pharmacy, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Zhenghao Xu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, No.548 Binwen Road, Hangzhou, Zhejiang, 310053, China.
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Chen F, Dong X, Wang Z, Wu T, Wei L, Li Y, Zhang K, Ma Z, Tian C, Li J, Zhao J, Zhang W, Liu A, Shen H. Regulation of specific abnormal calcium signals in the hippocampal CA1 and primary cortex M1 alleviates the progression of temporal lobe epilepsy. Neural Regen Res 2024; 19:425-433. [PMID: 37488907 PMCID: PMC10503629 DOI: 10.4103/1673-5374.379048] [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: 02/06/2023] [Revised: 03/15/2023] [Accepted: 05/04/2023] [Indexed: 07/26/2023] Open
Abstract
Temporal lobe epilepsy is a multifactorial neurological dysfunction syndrome that is refractory, resistant to antiepileptic drugs, and has a high recurrence rate. The pathogenesis of temporal lobe epilepsy is complex and is not fully understood. Intracellular calcium dynamics have been implicated in temporal lobe epilepsy. However, the effect of fluctuating calcium activity in CA1 pyramidal neurons on temporal lobe epilepsy is unknown, and no longitudinal studies have investigated calcium activity in pyramidal neurons in the hippocampal CA1 and primary motor cortex M1 of freely moving mice. In this study, we used a multi-channel fiber photometry system to continuously record calcium signals in CA1 and M1 during the temporal lobe epilepsy process. We found that calcium signals varied according to the grade of temporal lobe epilepsy episodes. In particular, cortical spreading depression, which has recently been frequently used to represent the continuously and substantially increased calcium signals, was found to correspond to complex and severe behavioral characteristics of temporal lobe epilepsy ranging from grade II to grade V. However, vigorous calcium oscillations and highly synchronized calcium signals in CA1 and M1 were strongly related to convulsive motor seizures. Chemogenetic inhibition of pyramidal neurons in CA1 significantly attenuated the amplitudes of the calcium signals corresponding to grade I episodes. In addition, the latency of cortical spreading depression was prolonged, and the above-mentioned abnormal calcium signals in CA1 and M1 were also significantly reduced. Intriguingly, it was possible to rescue the altered intracellular calcium dynamics. Via simultaneous analysis of calcium signals and epileptic behaviors, we found that the progression of temporal lobe epilepsy was alleviated when specific calcium signals were reduced, and that the end-point behaviors of temporal lobe epilepsy were improved. Our results indicate that the calcium dynamic between CA1 and M1 may reflect specific epileptic behaviors corresponding to different grades. Furthermore, the selective regulation of abnormal calcium signals in CA1 pyramidal neurons appears to effectively alleviate temporal lobe epilepsy, thereby providing a potential molecular mechanism for a new temporal lobe epilepsy diagnosis and treatment strategy.
Collapse
Affiliation(s)
- Feng Chen
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
- Institute for Translational Neuroscience, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xi Dong
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
- Institute for Translational Brain Research, Fudan University, Shanghai, China
| | - Zhenhuan Wang
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Tongrui Wu
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Liangpeng Wei
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
- Department of Radiology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuanyuan Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zengguang Ma
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Chao Tian
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingyu Zhao
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Wei Zhang
- Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, China
| | - Aili Liu
- Laboratory of Neurobiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hui Shen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Laboratory of Neurobiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Lawande NV, Conklin EA, Christian‐Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. Epilepsia Open 2023; 8:1512-1522. [PMID: 37715318 PMCID: PMC10690657 DOI: 10.1002/epi4.12828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing the main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. METHODS Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected ip, every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. RESULTS No differences in seizure susceptibility or mortality were observed between control males and control females. Gonadectomized mice exhibited increased susceptibility and reduced latency to both GS and SE in comparison to corresponding controls of the same sex, but the effects were stronger in males. In addition, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. SIGNIFICANCE The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | | | - Catherine A. Christian‐Hinman
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
5
|
Lawande NV, Conklin EA, Christian-Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541824. [PMID: 37292790 PMCID: PMC10245840 DOI: 10.1101/2023.05.22.541824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing a main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference, and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. Methods Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected i.p. every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. Results No differences in seizure susceptibility or mortality were observed between control males and control females. ORX males exhibited increased susceptibility and reduced latency to both GS and SE, but OVX females exhibited increased susceptibility and reduced latency to SE only. However, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. Significance The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Elisabeth A. Conklin
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Catherine A. Christian-Hinman
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| |
Collapse
|
6
|
Obydah W, Abouelnaga AF, Abass M, Saad S, Yehia A, Ammar OAA, Badawy AM, Ibrahim MM, Hussein AM. Possible Role of Oxidative Stress and Nrf2/HO-1 Pathway in Pentylenetetrazole-induced Epilepsy in Aged Rats. Rep Biochem Mol Biol 2023; 12:147-158. [PMID: 37724149 PMCID: PMC10505472 DOI: 10.52547/rbmb.12.1.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/14/2023] [Indexed: 09/20/2023]
Abstract
Background To examine the impact of aging on the response of rats to pentylenetetrazole (PTZ)-induction of epilepsy and the possible role of oxidative stress and nuclear factor erythroid 2-related factor 2 (Nrf2)/ heme oxygenase (HO)-1 pathway in this response. Methods Forty male albino rats were equally allocated into 4 groups; 1) Young control (YC) group, aged 8-12 weeks, 2) Old control (OC) group, aged 24 months, 3) PTZ-Young group: young rats received PTZ (50 mg/Kg, i.p. every other day) for 2 weeks and 4) PTZ-Old group: as group 3 but rats were old. The seizure score stage and latency to the first jerk were recorded in rats. Redox state markers in brain tissues including malondialdehyde (MDA), catalase and total antioxidant capacity (TAC) were evaluated. Also, the expression of Nrf2 and HO-1 genes were measured in the brain tissues. Results Old rats showed an early and a significant rise in the seizure score with PTZ administration and a significant drop in the seizure latency compared to young rats (P <0.01). Also, old rats showed a significantly higher MDA concentration and a significantly lower TAC and catalase activity than young rats (P <0.01). Moreover, the expression of Nrf2 and HO-1 was significantly lowered in old rats compared to young rats with PTZ administration (P < 0.01). Conclusion Aging increases the vulnerability of rats to PTZ-induced epilepsy. An effect might come down to the up-regulation of oxidative stress and the down regulation of antioxidant pathways including Nrf2 and HO-1.
Collapse
Affiliation(s)
- Walaa Obydah
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura (35516), Egypt.
| | - Ahmed Fathi Abouelnaga
- Department of Animal Husbandry and Development of animal wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Marwa Abass
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Somaya Saad
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura (35516), Egypt.
| | - Asmaa Yehia
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura (35516), Egypt.
| | | | - Alaa Mohamed Badawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Mohie Mahmoud Ibrahim
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Abdelaziz Mohamed Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura (35516), Egypt.
| |
Collapse
|
7
|
Boitet M, Eun H, Lee T, Kim J, Grailhe R. Non-invasive In Vivo Brain Astrogenesis and Astrogliosis Quantification Using a Far-red E2-Crimson Transgenic Reporter Mouse. Mol Neurobiol 2022; 59:6740-6753. [PMID: 36001234 DOI: 10.1007/s12035-022-02997-y] [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: 02/15/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
Abstract
Despite the adaptation of major clinical imaging modalities for small animals, optical bioluminescence imaging technology is the main approach readily reporting gene activity. Yet, in vivo bioluminescence monitoring requires the administration and diffusion of a substrate to the tissues of interest, resulting in experimental variability, high reagent cost, long acquisition time, and stress to the animal. In our study, we avoid such issues upon generating a new transgenic mouse (GFAP-E2crimson) expressing the far-red fluorescent protein E2-crimson under the control of the glial fibrillary acidic protein (GFAP) promoter. Using microscopy, we validated the selective expression of the reporter in the astrocyte cell population and by non-invasive in vivo fluorescence imaging its detection through the scalps and skulls of live animals. In addition, we performed a longitudinal study validating by in vivo imaging that the E2-crimson fluorescence signal is up-regulated, in pups during astrogenesis and in adult mice during astrogliosis upon kainic acid administration. Furthermore, upon crossing GFAP-E2crimson transgenic with 5XFAD Alzheimer's disease mice model, we were able to quantify the chronic inflammation triggered by amyloid deposit and aging over 18 months. As many diseases and conditions can trigger neuroinflammation, we believe that the GFAP-E2crimson reporter mice model delivers tremendous value for the non-invasive quantification of astrogliosis responses in living animals.
Collapse
Affiliation(s)
- Maylis Boitet
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
- Department of Biological Chemistry, IPK Campus, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeju Eun
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
| | - Taekwan Lee
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jiho Kim
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea
| | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea.
- Department of Biological Chemistry, IPK Campus, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
8
|
del Pozo A, Lehmann L, Knox KM, Barker-Haliski M. Can Old Animals Reveal New Targets? The Aging and Degenerating Brain as a New Precision Medicine Opportunity for Epilepsy. Front Neurol 2022; 13:833624. [PMID: 35572927 PMCID: PMC9096090 DOI: 10.3389/fneur.2022.833624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/07/2022] [Indexed: 02/02/2023] Open
Abstract
Older people represent the fastest growing group with epilepsy diagnosis. For example, cerebrovascular disease may underlie roughly 30-50% of epilepsy in older adults and seizures are also an underrecognized comorbidity of Alzheimer's disease (AD). As a result, up to 10% of nursing home residents may take antiseizure medicines (ASMs). Despite the greater incidence of epilepsy in older individuals and increased risk of comorbid seizures in people with AD, aged animals with seizures are strikingly underrepresented in epilepsy drug discovery practice. Increased integration of aged animals into preclinical epilepsy drug discovery could better inform the potential tolerability and pharmacokinetic interactions in aged individuals as the global population becomes increasingly older. Quite simply, the ASMs on the market today were brought forth based on efficacy in young adult, neurologically intact rodents; preclinical information concerning the efficacy and safety of promising ASMs is not routinely evaluated in aged animals. Integrating aged animals more often into basic epilepsy research may also uncover novel treatments for hyperexcitability. For example, cannabidiol and fenfluramine demonstrated clear efficacy in syndrome-specific pediatric models that led to a paradigm shift in the perceived value of pediatric models for ASM discovery practice; aged rodents with seizures or rodents with aging-related neuropathology represent an untapped resource that could similarly change epilepsy drug discovery. This review, therefore, summarizes how aged rodent models have thus far been used for epilepsy research, what studies have been conducted to assess ASM efficacy in aged rodent seizure and epilepsy models, and lastly to identify remaining gaps to engage aging-related neurological disease models for ASM discovery, which may simultaneously reveal novel mechanisms associated with epilepsy.
Collapse
Affiliation(s)
| | | | | | - Melissa Barker-Haliski
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| |
Collapse
|
9
|
Simeone KA, Wilke JC, Matthews SA, Simeone TA, Rho JM. Ketogenic diet-mediated seizure reduction preserves CA1 cell numbers in epileptic Kcna1-null mice: An unbiased stereological assessment. Epilepsia 2021; 62:e123-e128. [PMID: 34231878 DOI: 10.1111/epi.16983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
There is growing evidence for the disease-modifying potential of metabolic therapies, including the ketogenic diet (KD), which is used to treat medically intractable epilepsy. However, it remains unclear whether the KD exerts direct effects on histopathological changes in epileptic brain, or whether the changes are a consequence of diet-induced reduction in seizure activity. Here, we used unbiased stereological techniques to quantify the seizure-induced reduction in cell number in the CA1 region of the hippocampus of epileptic Kcna1-null mice and compared the effects of the KD with that of phenobarbital (PB), a widely employed anti-seizure drug. Our data suggest that the anti-seizure activity of the KD or PB was similar. However, CA1 cell numbers of KD-treated hippocampi were not significantly different from those seen in wild-type (WT) mice, whereas CA1 cell counts in standard diet and PB-treated Kcna1-null mice were 23% and 31% lower than WT animals, respectively. These results support the notion that structural protection of cells may involve more than seizure attenuation, and that the KD engages mechanisms that also promote or restore hippocampal morphological integrity.
Collapse
Affiliation(s)
- Kristina A Simeone
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Julianne C Wilke
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA.,Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Stephanie A Matthews
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Timothy A Simeone
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Jong M Rho
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA.,Rady Children's Hospital San Diego, San Diego, CA, USA.,Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
10
|
The Kainic Acid Models of Temporal Lobe Epilepsy. eNeuro 2021; 8:ENEURO.0337-20.2021. [PMID: 33658312 PMCID: PMC8174050 DOI: 10.1523/eneuro.0337-20.2021] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/14/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Experimental models of epilepsy are useful to identify potential mechanisms of epileptogenesis, seizure genesis, comorbidities, and treatment efficacy. The kainic acid (KA) model is one of the most commonly used. Several modes of administration of KA exist, each producing different effects in a strain-, species-, gender-, and age-dependent manner. In this review, we discuss the advantages and limitations of the various forms of KA administration (systemic, intrahippocampal, and intranasal), as well as the histologic, electrophysiological, and behavioral outcomes in different strains and species. We attempt a personal perspective and discuss areas where work is needed. The diversity of KA models and their outcomes offers researchers a rich palette of phenotypes, which may be relevant to specific traits found in patients with temporal lobe epilepsy.
Collapse
|
11
|
Lo AC, Rajan N, Gastaldo D, Telley L, Hilal ML, Buzzi A, Simonato M, Achsel T, Bagni C. Absence of RNA-binding protein FXR2P prevents prolonged phase of kainate-induced seizures. EMBO Rep 2021; 22:e51404. [PMID: 33779029 PMCID: PMC8024897 DOI: 10.15252/embr.202051404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.
Collapse
Affiliation(s)
- Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicholas Rajan
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Ludovic Telley
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Muna L Hilal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Buzzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Michele Simonato
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
12
|
Antagomir-mediated suppression of microRNA-134 reduces kainic acid-induced seizures in immature mice. Sci Rep 2021; 11:340. [PMID: 33431894 PMCID: PMC7801672 DOI: 10.1038/s41598-020-79350-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 12/04/2020] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are short non-coding RNAs that negatively regulate protein levels and perform important roles in establishing and maintaining neuronal network function. Previous studies in adult rodents have detected upregulation of microRNA-134 after prolonged seizures (status epilepticus) and demonstrated that silencing microRNA-134 using antisense oligonucleotides, termed antagomirs, has potent and long-lasting seizure-suppressive effects. Here we investigated whether targeting microRNA-134 can reduce or delay acute seizures in the immature brain. Status epilepticus was induced in 21 day-old (P21) male mice by systemic injection of 5 mg/kg kainic acid. This triggered prolonged electrographic seizures and select bilateral neuronal death within the CA3 subfield of the hippocampus. Expression of microRNA-134 and functional loading to Argonaute-2 was not significantly changed in the hippocampus after seizures in the model. Nevertheless, when levels of microRNA-134 were reduced by prior intracerebroventricular injection of an antagomir, kainic acid-induced seizures were delayed and less severe and mice displayed reduced neuronal death in the hippocampus. These studies demonstrate targeting microRNA-134 may have therapeutic applications for the treatment of seizures in children.
Collapse
|
13
|
Sun Y, Dhamne SC, Carretero-Guillén A, Salvador R, Goldenberg MC, Godlewski BR, Pascual-Leone A, Madsen JR, Stone SSD, Ruffini G, Márquez-Ruiz J, Rotenberg A. Drug-Responsive Inhomogeneous Cortical Modulation by Direct Current Stimulation. Ann Neurol 2020; 88:489-502. [PMID: 32542794 PMCID: PMC10675838 DOI: 10.1002/ana.25822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cathodal direct current stimulation (cDCS) induces long-term depression (LTD)-like reduction of cortical excitability (DCS-LTD), which has been tested in the treatment of epilepsy with modest effects. In part, this may be due to variable cortical neuron orientation relative to the electric field. We tested, in vivo and in vitro, whether DCS-LTD occurs throughout the cortical thickness, and if not, then whether drug-DCS pairing can enhance the uniformity of the cortical response and the cDCS antiepileptic effect. METHODS cDCS-mediated changes in cortical excitability were measured in vitro in mouse motor cortex (M1) and in human postoperative neocortex, in vivo in mouse somatosensory cortex (S1), and in a mouse kainic acid (KA)-seizure model. Contributions of N-methyl-D-aspartate-type glutamate receptors (NMDARs) to cDCS-mediated plasticity were tested with application of NMDAR blockers (memantine/D-AP5). RESULTS cDCS reliably induced DCS-LTD in superficial cortical layers, and a long-term potentiation (LTP)-like enhancement (DCS-LTP) was recorded in deep cortical layers. Immunostaining confirmed layer-specific increase of phospho-S6 ribosomal protein in mouse M1. Similar nonuniform cDCS aftereffects on cortical excitability were also found in human neocortex in vitro and in S1 of alert mice in vivo. Application of memantine/D-AP5 either produced a more uniform DCS-LTD throughout the cortical thickness or at least abolished DCS-LTP. Moreover, a combination of memantine and cDCS suppressed KA-induced seizures. INTERPRETATION cDCS aftereffects are not uniform throughout cortical layers, which may explain the incomplete cDCS clinical efficacy. NMDAR antagonists may augment cDCS efficacy in epilepsy and other disorders where regional depression of cortical excitability is desirable. ANN NEUROL 2020;88:489-502.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sameer C Dhamne
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Marti C Goldenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Repository Core, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Guttmann Institute, Autonomous University of Barcelona, Barcelona, Spain
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Scellig S D Stone
- Department of Neurosurgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Giulio Ruffini
- Neuroelectrics Corporation, Cambridge, Massachusetts, USA
| | - Javier Márquez-Ruiz
- Department of Physiology, Anatomy and Cellular Biology, Pablo de Olavide University, Seville, Spain
| | - Alexander Rotenberg
- Department of Neurology and the F. M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Guttmann Institute, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
14
|
Tam WY, Cheung KK. Phenotypic characteristics of commonly used inbred mouse strains. J Mol Med (Berl) 2020; 98:1215-1234. [PMID: 32712726 DOI: 10.1007/s00109-020-01953-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022]
Abstract
The laboratory mouse is the most commonly used mammalian model for biomedical research. An enormous number of mouse models, such as gene knockout, knockin, and overexpression transgenic mice, have been created over the years. A common practice to maintain a genetically modified mouse line is backcrossing with standard inbred mice over several generations. However, the choice of inbred mouse for backcrossing is critical to phenotypic characterization because phenotypic variabilities are often observed between mice with different genetic backgrounds. In this review, the major features of commonly used inbred mouse lines are discussed. The aim is to provide information for appropriate selection of inbred mouse lines for genetic and behavioral studies.
Collapse
Affiliation(s)
- Wing Yip Tam
- University Research Facility in Behavioral and Systems Neuroscience, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Kwok-Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| |
Collapse
|
15
|
Armas-Capote N, Maglio LE, Pérez-Atencio L, Martin-Batista E, Reboreda A, Barios JA, Hernandez G, Alvarez de la Rosa D, Lamas JA, Barrio LC, Giraldez T. SGK1.1 Reduces Kainic Acid-Induced Seizure Severity and Leads to Rapid Termination of Seizures. Cereb Cortex 2019; 30:3184-3197. [DOI: 10.1093/cercor/bhz302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/19/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
Abstract
Approaches to control epilepsy, one of the most important idiopathic brain disorders, are of great importance for public health. We have previously shown that in sympathetic neurons the neuronal isoform of the serum and glucocorticoid-regulated kinase (SGK1.1) increases the M-current, a well-known target for seizure control. The effect of SGK1.1 activation on kainate-induced seizures and neuronal excitability was studied in transgenic mice that express a permanently active form of the kinase, using electroencephalogram recordings and electrophysiological measurements in hippocampal brain slices. Our results demonstrate that SGK1.1 activation leads to reduced seizure severity and lower mortality rates following status epilepticus, in an M-current–dependent manner. EEG is characterized by reduced number, shorter duration, and early termination of kainate-induced seizures in the hippocampus and cortex. Hippocampal neurons show decreased excitability associated to increased M-current, without altering basal synaptic transmission or other neuronal properties. Altogether, our results reveal a novel and selective anticonvulsant pathway that promptly terminates seizures, suggesting that SGK1.1 activation can be a potent factor to secure the brain against permanent neuronal damage associated to epilepsy.
Collapse
Affiliation(s)
- Natalia Armas-Capote
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| | - Laura E Maglio
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| | - Leonel Pérez-Atencio
- Unidad de Neurologia Experimental, Hospital Ramon y Cajal-IRYCIS, Madrid, 28034 Spain
| | - Elva Martin-Batista
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| | - Antonio Reboreda
- Department of Functional Biology and Health Sciences, Faculty of Biology-CINBIO-IBIV, University of Vigo, Vigo, 36310 Spain
| | - Juan A Barios
- Systems Engineering and Automation Department, Miguel Hernandez University, Elche, 03202 Spain
| | - Guadalberto Hernandez
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| | - José Antonio Lamas
- Department of Functional Biology and Health Sciences, Faculty of Biology-CINBIO-IBIV, University of Vigo, Vigo, 36310 Spain
| | - Luis C Barrio
- Unidad de Neurologia Experimental, Hospital Ramon y Cajal-IRYCIS, Madrid, 28034 Spain
| | - Teresa Giraldez
- Departamento de Ciencias Medicas Basicas-Fisiologia and Instituto de Tecnologías Biomedicas (ITB), Universidad de La Laguna, Tenerife, 38071 Spain
| |
Collapse
|
16
|
Maciąg F, Majewski Ł, Boguszewski PM, Gupta RK, Wasilewska I, Wojtaś B, Kuznicki J. Behavioral and electrophysiological changes in female mice overexpressing ORAI1 in neurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1137-1150. [PMID: 30659848 DOI: 10.1016/j.bbamcr.2019.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Orai proteins form highly selective Ca2+ release-activated channels (CRACs). They play a critical role in store-operated Ca2+ entry (SOCE; i.e., the influx of external Ca2+ that is induced by the depletion of endoplasmic reticulum Ca2+ stores). Of the three Orai homologs that are present in mammals (Orai1-3), the physiological function of Orai1 is the best described. CRACs are formed by both homomeric assemblies and heteromultimers of Orais. Orai1 and Orai2 can form heteromeric channels that differ in conductivity during SOCE, depending on their Orai1-to-Orai2 ratio. The present study explored the potential consequences of ORAI1 overexpression in neurons where the dominant isoform is Orai2. We established the Tg(ORAI1)Ibd transgenic mouse line that overexpresses ORAI1 in brain neurons. We observed seizure-like symptoms in aged (≥15-month-old) female mice but not in males of the same age. The application of kainic acid and bicuculline to slices that were isolated from 8-month-old (±1 month) female Tg(ORAI1)Ibd mice revealed a significantly lower frequency of interictal bursts compared with samples that were isolated from wildtype mice. No differences were observed in male mice of a similar age. A battery of behavioral tests showed that context recognition decreased only in female transgenic mice. The phenotype that was observed in female mice suggests that ORAI1 overexpression may affect neuronal activity in a sex-dependent manner. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Filip Maciąg
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., Warsaw 02-109, Poland
| | - Łukasz Majewski
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., Warsaw 02-109, Poland.
| | - Paweł M Boguszewski
- Laboratory of Animal Models, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
| | - Rishikesh Kumar Gupta
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., Warsaw 02-109, Poland
| | - Iga Wasilewska
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., Warsaw 02-109, Poland
| | - Bartosz Wojtaś
- Laboratory of Molecular Neurobiology, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Jacek Kuznicki
- International Institute of Molecular and Cell Biology in Warsaw, 4 Ks. Trojdena Str., Warsaw 02-109, Poland
| |
Collapse
|
17
|
Jin Z, Jung Y, Yi CO, Lee JY, Jeong EA, Lee JE, Park KJ, Kwon OY, Lim BH, Choi NC, Roh GS. Atorvastatin pretreatment attenuates kainic acid-induced hippocampal neuronal death via regulation of lipocalin-2-associated neuroinflammation. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:301-309. [PMID: 29719452 PMCID: PMC5928343 DOI: 10.4196/kjpp.2018.22.3.301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/31/2017] [Accepted: 02/07/2018] [Indexed: 11/15/2022]
Abstract
Statins mediate vascular protection and reduce the prevalence of cardiovascular diseases. Recent work indicates that statins have anticonvulsive effects in the brain; however, little is known about the precise mechanism for its protective effect in kainic acid (KA)-induced seizures. Here, we investigated the protective effects of atorvastatin pretreatment on KA-induced neuroinflammation and hippocampal cell death. Mice were treated via intragastric administration of atorvastatin for 7 days, injected with KA, and then sacrificed after 24 h. We observed that atorvastatin pretreatment reduced KA-induced seizure activity, hippocampal cell death, and neuroinflammation. Atorvastatin pretreatment also inhibited KA-induced lipocalin-2 expression in the hippocampus and attenuated KA-induced hippocampal cyclooxygenase-2 expression and glial activation. Moreover, AKT phosphorylation in KA-treated hippocampus was inhibited by atorvastatin pretreatment. These findings suggest that atorvastatin pretreatment may protect hippocampal neurons during seizures by controlling lipocalin-2-associated neuroinflammation.
Collapse
Affiliation(s)
- Zhen Jin
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Yohan Jung
- Department of Neurology, Changwon Fatima Hospital, Changwon 51394, Korea
| | - Chin-Ok Yi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Eun Ae Jeong
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Jung Eun Lee
- Department of Thoracic and Cardiovascular Surgery, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Ki-Jong Park
- Department of Neurology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Oh-Young Kwon
- Department of Neurology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Byeong Hoon Lim
- Department of Neurology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Nack-Cheon Choi
- Department of Neurology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
18
|
Dubey M, Brouwers E, Hamilton EM, Stiedl O, Bugiani M, Koch H, Kole MH, Boschert U, Wykes RC, Mansvelder HD, van der Knaap MS, Min R. Seizures and disturbed brain potassium dynamics in the leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts. Ann Neurol 2018; 83:636-649. [PMID: 29466841 PMCID: PMC5900999 DOI: 10.1002/ana.25190] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 01/12/2018] [Accepted: 02/18/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Loss of function of the astrocyte-specific protein MLC1 leads to the childhood-onset leukodystrophy "megalencephalic leukoencephalopathy with subcortical cysts" (MLC). Studies on isolated cells show a role for MLC1 in astrocyte volume regulation and suggest that disturbed brain ion and water homeostasis is central to the disease. Excitability of neuronal networks is particularly sensitive to ion and water homeostasis. In line with this, reports of seizures and epilepsy in MLC patients exist. However, systematic assessment and mechanistic understanding of seizures in MLC are lacking. METHODS We analyzed an MLC patient inventory to study occurrence of seizures in MLC. We used two distinct genetic mouse models of MLC to further study epileptiform activity and seizure threshold through wireless extracellular field potential recordings. Whole-cell patch-clamp recordings and K+ -sensitive electrode recordings in mouse brain slices were used to explore the underlying mechanisms of epilepsy in MLC. RESULTS An early onset of seizures is common in MLC. Similarly, in MLC mice, we uncovered spontaneous epileptiform brain activity and a lowered threshold for induced seizures. At the cellular level, we found that although passive and active properties of individual pyramidal neurons are unchanged, extracellular K+ dynamics and neuronal network activity are abnormal in MLC mice. INTERPRETATION Disturbed astrocyte regulation of ion and water homeostasis in MLC causes hyperexcitability of neuronal networks and seizures. These findings suggest a role for defective astrocyte volume regulation in epilepsy. Ann Neurol 2018;83:636-649.
Collapse
Affiliation(s)
- Mohit Dubey
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
- Present address:
Current address for Mohit Dubey: Department of Axonal SignalingNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Eelke Brouwers
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
| | - Eline M.C. Hamilton
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
| | - Oliver Stiedl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
| | - Marianna Bugiani
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
- Department of PathologyVU University Medical CenterAmsterdamThe Netherlands
| | - Henner Koch
- Department of NeurologyUniversity of Tübingen, Hertie Institute for Clinical Brain ResearchTübingenGermany
| | - Maarten H.P. Kole
- Department of Axonal SignalingNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
- Cell Biology, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Ursula Boschert
- Translational Innovation Platform Immunology/Neurology, EMD Serono Research & Development InstituteBillericaMA
| | - Robert C. Wykes
- Department of Clinical & Experimental Epilepsy, UCL Institute of NeurologyUniversity College LondonLondonUnited Kingdom
| | - Huibert D. Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam NeuroscienceVU University Medical CenterAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVU UniversityAmsterdamThe Netherlands
| |
Collapse
|
19
|
Abstract
Since its discovery the cellular prion protein (encoded by the Prnp gene) has been associated with a large number of functions. The proposed functions rank from basic cellular processes such as cell cycle and survival to neural functions such as behavior and neuroprotection, following a pattern similar to that of Moore's law for electronics. In addition, particular interest is increasing in the participation of Prnp in neurodegeneration. However, in recent years a redefinition of these functions has begun, since examples of previously attributed functions were increasingly re-associated with other proteins. Most of these functions are linked to so-called "Prnp-flanking genes" that are close to the genomic locus of Prnp and which are present in the genome of some Prnp mouse models. In addition, their role in neuroprotection against convulsive insults has been confirmed in recent studies. Lastly, in recent years a large number of models indicating the participation of different domains of the protein in apoptosis have been uncovered. However, after more than 10 years of molecular dissection our view is that the simplest mechanistic model in PrP(C)-mediated cell death should be considered, as Ockham's razor theory suggested.
Collapse
Affiliation(s)
- José A del Río
- a Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC) , Parc Científic de Barcelona, Barcelona , Spain.,b Department of Cell Biology, Physiology and Inmunology , Facultat de Biologia, Universitat de Barcelona , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Barcelona , Spain
| | - Rosalina Gavín
- a Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC) , Parc Científic de Barcelona, Barcelona , Spain.,b Department of Cell Biology, Physiology and Inmunology , Facultat de Biologia, Universitat de Barcelona , Barcelona , Spain.,c Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Barcelona , Spain
| |
Collapse
|
20
|
Kohnken RA, Schwahn DJ. Lack of Chronic Histologic Lesions Supportive of Sublethal Spontaneous Seizures in FVB/N Mice. Comp Med 2016; 66:105-11. [PMID: 27053564 PMCID: PMC4825959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/21/2015] [Accepted: 09/04/2015] [Indexed: 06/05/2023]
Abstract
FVB/N mice with 'space cadet' syndrome are prone to audiogenic seizures and are considered excitotoxic 'sensitive' mice due to the neuronal damage that accompanies seizures. FVB/N mice found dead demonstrate acute neuronal cell death--attributed to a massive seizure episode--within the hippocampus and cerebrocortical laminae. However, the behavioral features of FVB/N mice and numerous studies using excitotoxins to induce seizure activity indicate that this strain experiences multiple sublethal seizures. To assess whether FVB/N mice develop histologically detectable lesions, we evaluated the brains of 86 aged (154-847 d) FVB/N mice without a history of seizures. The hippocampus and cerebrocortical laminae were evaluated histologically for neuronal atrophy and gliosis. Neuronal atrophy was quantified by counting neurons in the hippocampus (CA3 and dentate gyrus) and cerebral cortex. Gliosis was quantified by using immunohistochemistry for glial fibrillary acidic protein and glial counting in the cerebral cortex. In addition, ventricular area was calculated. Our study revealed no changes in brain weight with age, no neuronal loss or gliosis, no correlation between neuronal or glial cell profile densities and brain weight or age, and no differences in ventricular size between FVB/N and control mice. Neuronal densities in the cerebral cortex and granule cells of the dentate gyrus were lower in FVB/N mice than in control Swiss Webster mice. We conclude that although acute lesions of seizure activity are a previous feature of the FVB/N strain, chronic seizure activity in these mice either is negligible or does not cause morphologic or phenotypic changes.
Collapse
Affiliation(s)
- Rebecca A Kohnken
- Department of Veterinary Biosciences, Ohio State University, Columbus, Ohio, USA
| | - Denise J Schwahn
- Research Animal Resources Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
21
|
Umpierre AD, Bennett IV, Nebeker LD, Newell TG, Tian BB, Thomson KE, White HS, White JA, Wilcox KS. Repeated low-dose kainate administration in C57BL/6J mice produces temporal lobe epilepsy pathology but infrequent spontaneous seizures. Exp Neurol 2016; 279:116-126. [PMID: 26896834 DOI: 10.1016/j.expneurol.2016.02.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/15/2016] [Indexed: 12/31/2022]
Abstract
More efficient or translationally relevant approaches are needed to model acquired temporal lobe epilepsy (TLE) in genetically tractable mice. The high costs associated with breeding and maintaining transgenic, knock-in, or knock-out lines place a high value on the efficiency of induction and animal survivability. Herein, we describe our approaches to model acquired epilepsy in C57BL/6J mice using repeated, low-dose kainate (KA) administration paradigms. Four paradigms (i.p.) were tested for their ability to induce status epilepticus (SE), temporal lobe pathology, and the development of epilepsy. All four paradigms reliably induce behavioral and/or electrographic SE without mortality over a 7d period. Two of the four paradigms investigated produce features indicative of TLE pathology, including hippocampal cell death, widespread astrogliosis, and astrocyte expression of mGluR5, a feature commonly reported in TLE models. Three of the investigated paradigms were able to produce aberrant electrographic features, such as interictal spiking in cortex. However, only one paradigm, previously published by others, produces spontaneous recurrent seizures over an eight week period. Presentation of spontaneous seizures is rare (N=2/14), with epilepsy preferentially developing in animals having a high number of seizures during SE. Overall, repeated, low-dose KA administration improves the efficiency and pathological relevance of a systemic KA insult, but does not produce a robust epilepsy phenotype under the experimental paradigms described herein.
Collapse
Affiliation(s)
- Anthony D Umpierre
- Interdepartmental Program in Neuroscience, University of Utah, 401 MREB, 20N. 1900E, Salt Lake City, UT 84132, United States.
| | - Isaiah V Bennett
- Dept. of Pharmacology & Toxicology, University of Utah, College of Pharmacy, 30N. 1900E, Salt Lake City, UT 84112, United States.
| | - Lismore D Nebeker
- Dept. of Pharmacology & Toxicology, University of Utah, College of Pharmacy, 30N. 1900E, Salt Lake City, UT 84112, United States.
| | - Thomas G Newell
- Anticonvulsant Drug Development Program, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, United States; Dept. of Bioengineering, University of Utah, 36S. Wasatch Drive, Rm. 3100, Salt Lake City, UT 84112, United States.
| | - Bruce B Tian
- Dept. of Pharmacology & Toxicology, University of Utah, College of Pharmacy, 30N. 1900E, Salt Lake City, UT 84112, United States.
| | - Kyle E Thomson
- Anticonvulsant Drug Development Program, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, United States; Dept. of Bioengineering, University of Utah, 36S. Wasatch Drive, Rm. 3100, Salt Lake City, UT 84112, United States.
| | - H Steve White
- Dept. of Pharmacology & Toxicology, University of Utah, College of Pharmacy, 30N. 1900E, Salt Lake City, UT 84112, United States; Anticonvulsant Drug Development Program, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, United States; Department of Pharmacy, University of Washington, Box 357630, H375 Health Science Building, Seattle, WA 98195, United States.
| | - John A White
- Dept. of Biomedical Engineering, Boston University, 44 Cummington Mall, Rm. 403, Boston, MA 02215, United States.
| | - Karen S Wilcox
- Interdepartmental Program in Neuroscience, University of Utah, 401 MREB, 20N. 1900E, Salt Lake City, UT 84132, United States; Dept. of Pharmacology & Toxicology, University of Utah, College of Pharmacy, 30N. 1900E, Salt Lake City, UT 84112, United States; Anticonvulsant Drug Development Program, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, United States.
| |
Collapse
|
22
|
Immediate Epileptogenesis after Kainate-Induced Status Epilepticus in C57BL/6J Mice: Evidence from Long Term Continuous Video-EEG Telemetry. PLoS One 2015; 10:e0131705. [PMID: 26161754 PMCID: PMC4498886 DOI: 10.1371/journal.pone.0131705] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/04/2015] [Indexed: 11/19/2022] Open
Abstract
The C57BL/6J mouse as a model of seizure/epilepsy is challenging due to high mortality and huge variability in response to kainate. We have recently demonstrated that repeated administration of a low dose of kainate by intraperitoneal route can induce severe status epilepticus (SE) with 94% survival rate. In the present study, based on continuous video-EEG recording for 4-18 weeks from epidurally implanted electrodes on the cortex, we demonstrate that this method also induces immediate epileptogenesis (<1-5 days post-SE). This finding was based on identification of two types of spontaneous recurrent seizures; behavioral convulsive seizures (CS) and electrographic nonconvulsive seizures (NCS). The identification of the spontaneous CS, stage 3-5 types, was based on the behaviors (video) that were associated with the EEG characteristics (stage 3-5 epileptiform spikes), the power spectrum, and the activity counts. The electrographic NCS identification was based on the stage 1-2 epileptiform spike clusters on the EEG and their associated power spectrum. Severe SE induced immediate epileptogenesis in all the mice. The maximum numbers of spontaneous CS were observed during the first 4-6 weeks of the SE and they decreased thereafter. Mild SE also induced immediate epileptogenesis in some mice but the CS were less frequent. In both the severe and the mild SE groups, the spontaneous electrographic NCS persisted throughout the 18 weeks observation period, and therefore this could serve as a chronic model for complex seizures. However, unlike rat kainate models, the C57BL/6J mouse kainate model is a unique regressive CS model of epilepsy. Further studies are required to understand the mechanism of recovery from spontaneous CS in this model, which could reveal novel therapeutic targets for epilepsy.
Collapse
|
23
|
Carulla P, Llorens F, Matamoros-Angles A, Aguilar-Calvo P, Espinosa JC, Gavín R, Ferrer I, Legname G, Torres JM, del Río JA. Involvement of PrP(C) in kainate-induced excitotoxicity in several mouse strains. Sci Rep 2015; 5:11971. [PMID: 26155834 PMCID: PMC4648388 DOI: 10.1038/srep11971] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/12/2015] [Indexed: 01/14/2023] Open
Abstract
The cellular prion protein (PrP(C)) has been associated with a plethora of cellular functions ranging from cell cycle to neuroprotection. Mice lacking PrP(C) show an increased susceptibility to epileptic seizures; the protein, then, is neuroprotective. However, lack of experimental reproducibility has led to considering the possibility that other factors besides PrP(C) deletion, such as the genetic background of mice or the presence of so-called "Prnp flanking genes", might contribute to the reported susceptibility. Here, we performed a comparative analysis of seizure-susceptibility using characterized Prnp(+/+) and Prnp(0/0) mice of B6129, B6.129, 129/Ola or FVB/N genetic backgrounds. Our study indicates that PrP(C) plays a role in neuroprotection in KA-treated cells and mice. For this function, PrP(C) should contain the aa32-93 region and needs to be linked to the membrane. In addition, some unidentified "Prnp-flanking genes" play a role parallel to PrP(C) in the KA-mediated responses in B6129 and B6.129 Prnp(0/0) mice.
Collapse
Affiliation(s)
- Patricia Carulla
- 1] Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain [2] Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain [3] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Franc Llorens
- 1] Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain [2] Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain [3] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain [4] German Center for Neurodegenerative Diseases (DZNE), Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - Andreu Matamoros-Angles
- 1] Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain [2] Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain [3] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | | | - Juan Carlos Espinosa
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Rosalina Gavín
- 1] Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain [2] Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain [3] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Isidre Ferrer
- 1] Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Juan Maria Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - José A del Río
- 1] Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain [2] Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain [3] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| |
Collapse
|
24
|
Mitochondrial ATP-Mg/Pi carrier SCaMC-3/Slc25a23 counteracts PARP-1-dependent fall in mitochondrial ATP caused by excitotoxic insults in neurons. J Neurosci 2015; 35:3566-81. [PMID: 25716855 DOI: 10.1523/jneurosci.2702-14.2015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Glutamate excitotoxicity is caused by sustained activation of neuronal NMDA receptors causing a large Ca(2+) and Na(+) influx, activation of poly(ADP ribose) polymerase-1 (PARP-1), and delayed Ca(2+) deregulation. Mitochondria undergo early changes in membrane potential during excitotoxicity, but their precise role in these events is still controversial. Using primary cortical neurons derived from mice, we show that NMDA exposure results in a rapid fall in mitochondrial ATP in neurons deficient in SCaMC-3/Slc25a23, a Ca(2+)-regulated mitochondrial ATP-Mg/Pi carrier. This fall is associated with blunted increases in respiration and a delayed decrease in cytosolic ATP levels, which are prevented by PARP-1 inhibitors or by SCaMC-3 activity promoting adenine nucleotide uptake into mitochondria. SCaMC-3 KO neurons show an earlier delayed Ca(2+) deregulation, and SCaMC-3-deficient mitochondria incubated with ADP or ATP-Mg had reduced Ca(2+) retention capacity, suggesting a failure to maintain matrix adenine nucleotides as a cause for premature delayed Ca(2+) deregulation. SCaMC-3 KO neurons have higher vulnerability to in vitro excitotoxicity, and SCaMC-3 KO mice are more susceptible to kainate-induced seizures, showing that early PARP-1-dependent fall in mitochondrial ATP levels, counteracted by SCaMC-3, is an early step in the excitotoxic cascade.
Collapse
|
25
|
Kim TY, Yoshimoto T, Aoyama Y, Niimi K, Takahashi E, Itakura C. Age-dependent kainate sensitivity in heterozygous rolling Nagoya Cav2.1 channel mutant mice. Pharmacol Biochem Behav 2014; 124:250-9. [DOI: 10.1016/j.pbb.2014.06.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 12/29/2022]
|
26
|
Hui Yin Y, Ahmad N, Makmor-Bakry M. Pathogenesis of epilepsy: challenges in animal models. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2013; 16:1119-32. [PMID: 24494063 PMCID: PMC3909622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/02/2013] [Indexed: 11/13/2022]
Abstract
Epilepsy is one of the most common chronic disorders affecting individuals of all ages. A greater understanding of pathogenesis in epilepsy will likely provide the basis fundamental for development of new antiepileptic therapies that aim to prevent the epileptogenesis process or modify the progression of epilepsy in addition to treatment of epilepsy symptomatically. Therefore, several investigations have embarked on advancing knowledge of the mechanism underlying epileptogenesis, understanding in mechanism of pharmacoresistance and discovering antiepileptogenic or disease-modifying therapy. Animal models play a crucial and significant role in providing additional insight into mechanism of epileptogenesis. With the help of these models, epileptogenesis process has been demonstrated to be involved in various molecular and biological pathways or processes. Hence, this article will discuss the known and postulated mechanisms of epileptogenesis and challenges in using the animal models.
Collapse
Affiliation(s)
- Yow Hui Yin
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurulumi Ahmad
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Makmor-Bakry
- Faculty of Pharmacy, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia,Corresponding author: Mohd Makmor-Bakry. Faculty of Pharmacy, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia. Tel: +60392897244; Fax: +60326983271;
| |
Collapse
|
27
|
Striebel JF, Race B, Pathmajeyan M, Rangel A, Chesebro B. Lack of influence of prion protein gene expression on kainate-induced seizures in mice: studies using congenic, coisogenic and transgenic strains. Neuroscience 2013; 238:11-8. [PMID: 23415788 PMCID: PMC3676307 DOI: 10.1016/j.neuroscience.2013.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/17/2013] [Accepted: 02/01/2013] [Indexed: 01/09/2023]
Abstract
Prion protein (PrP) is a glycosylphosphatidylinositol (GPI) anchored cell surface protein expressed by many cells, including those of the mammalian nervous system. At present the physiologic functions of PrP remain unclear. Deletion of Prnp, the gene encoding PrP in mice, has been shown to alter normal synaptic and electrophysiologic activities, indicating a potential role in seizure susceptibility. However, published efforts to link PrP with seizures, using both in vivo and in vitro models, are conflicting and difficult to interpret due to use of various mouse backgrounds and seizure induction techniques. Here we investigated the role of PrP in kainic acid (KA)-induced seizure sensitivity, using three types of mice. In contrast to previous published results, Prnp-/- mice on the C57BL/10SnJ background had a significant decrease in KA-induced seizure susceptibility. In genetic complementation experiments using a PrP-expressing transgene, genes derived from strain 129/Ola, which flanked the Prnp-/- locus in C57BL/10SnJ mice, rather than Prnp itself, appeared to account for this effect. Furthermore, using coisogenic 129/Ola mice differing only at Prnp, this difference was not reproduced when comparing PrP-negative and PrP-positive mice. In contrast, substrains of PrP-expressing C57BL mice, showed large variations in KA-induced seizure sensitivity. The magnitude of these differences in susceptibility was larger than that associated with the presence of the Prnp gene, suggesting extensive influence of genes other than Prnp on seizure sensitivity in this system.
Collapse
Affiliation(s)
- James F. Striebel
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Brent Race
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Melissa Pathmajeyan
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Alejandra Rangel
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Bruce Chesebro
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| |
Collapse
|
28
|
The neuronal serum- and glucocorticoid-regulated kinase 1.1 reduces neuronal excitability and protects against seizures through upregulation of the M-current. J Neurosci 2013; 33:2684-96. [PMID: 23392695 DOI: 10.1523/jneurosci.3442-12.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The M-current formed by tetramerization of Kv7.2 and Kv7.3 subunits is a neuronal voltage-gated K(+) conductance that controls resting membrane potential and cell excitability. In Xenopus laevis oocytes, an increase in Kv7.2/3 function by the serum- and glucocorticoid-regulated kinase 1 (SGK1) has been reported previously (Schuetz et al., 2008). We now show that the neuronal isoform of this kinase (SGK1.1), with distinct subcellular localization and modulation, upregulates the Kv7.2/3 current in Xenopus oocytes and mammalian human embryonic kidney HEK293 cells. In contrast to the ubiquitously expressed SGK1, the neuronal isoform SGK1.1 interacts with phosphoinositide-phosphatidylinositol 4,5-bisphosphate (PIP(2)) and is distinctly localized to the plasma membrane (Arteaga et al., 2008). An SGK1.1 mutant with disrupted PIP(2) binding sites produced no effect on Kv7.2/3 current amplitude. SGK1.1 failed to modify the voltage dependence of activation and did not change activation or deactivation kinetics of Kv7.2/3 channels. These results suggest that the kinase increases channel membrane abundance, which was confirmed with flow cytometry assays. To evaluate the effect of the kinase in neuronal excitability, we generated a transgenic mouse (Tg.sgk) expressing a constitutively active form of SGK1.1 (S515D). Superior cervical ganglion (SCG) neurons isolated from Tg.sgk mice showed a significant increase in M-current levels, paralleled by reduced excitability and more negative resting potentials. SGK1.1 effect on M-current in Tg.sgk-SCG neurons was counteracted by muscarinic receptor activation. Transgenic mice with increased SGK1.1 activity also showed diminished sensitivity to kainic acid-induced seizures. Altogether, our results unveil a novel role of SGK1.1 as a physiological regulator of the M-current and neuronal excitability.
Collapse
|
29
|
Zhu XB, Wang YB, Chen O, Zhang DQ, Zhang ZH, Cao AH, Huang SY, Sun RP. Characterization of the expression of macrophage inflammatory protein-1α (MIP-1α) and C-C chemokine receptor 5 (CCR5) after kainic acid-induced status epilepticus (SE) in juvenile rats. Neuropathol Appl Neurobiol 2012; 38:602-16. [DOI: 10.1111/j.1365-2990.2012.01251.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Cho E, Lee KJ, Seo JW, Byun CJ, Chung SJ, Suh DC, Carmeliet P, Koh JY, Kim JS, Lee JY. Neuroprotection by urokinase plasminogen activator in the hippocampus. Neurobiol Dis 2012; 46:215-24. [PMID: 22293605 DOI: 10.1016/j.nbd.2012.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 01/01/2023] Open
Abstract
Tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA), which are both used for thrombolytic treatment of acute ischemic stroke, are serine proteases that convert plasminogen to active plasmin. Although recent experimental evidences have raised controversy about the neurotoxic versus neuroprotective roles of tPA in acute brain injury, uPA remains unexplored in this context. In this study, we evaluated the effect of uPA on neuronal death in the hippocampus of mice after kainate-induced seizures. In the normal brain, uPA was localized to both nuclei and cytosol of neurons. Following severe kainate-induced seizures, uPA completely disappeared in degenerating neurons, whereas uPA-expressing astrocytes substantially increased, suggesting reactive astrogliosis. uPA-knockout mice were more vulnerable to kainate-induced neuronal death than wild-type mice. Consistent with this, inhibition of uPA by intracerebral injection of the uPA inhibitor UK122 increased the level of neuronal death. In contrast, prior administration of recombinant uPA significantly attenuated neuronal death. Collectively, these results indicate that uPA renders neurons resistant to kainate-induced excitotoxicity. Moreover, recombinant uPA suppressed cell death in primary cultures of hippocampal neurons exposed to H2O2, zinc, or various excitotoxins, suggesting that uPA protects against neuronal injuries mediated by the glutamate receptor, or by oxidation- or zinc-induced death signaling pathways. Considering that tPA may facilitate neurodegeneration in acute brain injury, we suggest that uPA, as a neuroprotectant, might be beneficial for the treatment of acute brain injuries such as ischemic stroke.
Collapse
Affiliation(s)
- Eunsil Cho
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kuruba R, Hattiangady B, Parihar VK, Shuai B, Shetty AK. Differential susceptibility of interneurons expressing neuropeptide Y or parvalbumin in the aged hippocampus to acute seizure activity. PLoS One 2011; 6:e24493. [PMID: 21915341 PMCID: PMC3167860 DOI: 10.1371/journal.pone.0024493] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 08/10/2011] [Indexed: 12/28/2022] Open
Abstract
Acute seizure (AS) activity in old age has an increased predisposition for evolving into temporal lobe epilepsy (TLE). Furthermore, spontaneous seizures and cognitive dysfunction after AS activity are often intense in the aged population than in young adults. This could be due to an increased vulnerability of inhibitory interneurons in the aged hippocampus to AS activity. We investigated this issue by comparing the survival of hippocampal GABA-ergic interneurons that contain the neuropeptide Y (NPY) or the calcium binding protein parvalbumin (PV) between young adult (5-months old) and aged (22-months old) F344 rats at 12 days after three-hours of AS activity. Graded intraperitoneal injections of the kainic acid (KA) induced AS activity and a diazepam injection at 3 hours after the onset terminated AS-activity. Measurement of interneuron numbers in different hippocampal subfields revealed that NPY+ interneurons were relatively resistant to AS activity in the aged hippocampus in comparison to the young adult hippocampus. Whereas, PV+ interneurons were highly susceptible to AS activity in both age groups. However, as aging alone substantially depleted these populations, the aged hippocampus after three-hours of AS activity exhibited 48% reductions in NPY+ interneurons and 70% reductions in PV+ interneurons, in comparison to the young hippocampus after similar AS activity. Thus, AS activity-induced TLE in old age is associated with far fewer hippocampal NPY+ and PV+ interneuron numbers than AS-induced TLE in the young adult age. This discrepancy likely underlies the severe spontaneous seizures and cognitive dysfunction observed in the aged people after AS activity.
Collapse
Affiliation(s)
- Ramkumar Kuruba
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bharathi Hattiangady
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- Institute for Regenerative Medicine, Texas A&M Health Science Center at Scott & White, Temple, Texas, United States of America
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Vipan K. Parihar
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bing Shuai
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- Institute for Regenerative Medicine, Texas A&M Health Science Center at Scott & White, Temple, Texas, United States of America
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ashok K. Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, North Carolina, United States of America
- Institute for Regenerative Medicine, Texas A&M Health Science Center at Scott & White, Temple, Texas, United States of America
- Research Service, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Zhang K, Tolstykh GP, Sanchez RM, Cavazos JE. Chronic Cellular Hyperexcitability in Elderly Epileptic Rats with Spontaneous Seizures Induced by Kainic Acid Status Epilepticus while Young Adults. Aging Dis 2011; 2:332-338. [PMID: 22396885 PMCID: PMC3295074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2011] [Revised: 08/22/2011] [Accepted: 08/22/2011] [Indexed: 05/31/2023] Open
Abstract
Emerging data indicate that age-related brain changes alter seizure susceptibility, seizure-associated neurodegeneration, and responsiveness to AEDs. The present study assessed long-term animal survival in the Kainic Acid (KA) model along with in-vivo spontaneous seizure frequency, cellular hyperexcitability in CA1 in-vitro and in-vivo in subiculum, and responsiveness of in-vitro CA1 hyperexcitability to topiramate. Sprague-Dawley male rats were given KA to induce convulsive status epilepticus (KA-SE) at 2-3 months of age. The one-month mortality after KA-SE was 27%. One-month survivor rats had 37% sudden unexplained late mortality after KA-SE as compared to none in saline controls during their second year of life. In-vivo seizure frequency was examined prior to terminal experiments. The diurnal average seizure frequency in the KA-SE group at age 2 years was 1.06 ± 0.24 seizures/hour while no seizures were observed in the saline age-matched controls (p<0.001). In-vitro recordings of CA1 pyramidal neurons revealed that depolarizing current injection from -60 mV evoked an increased number of action potentials in the aged KA-SE group compared to controls (p<0.002). Topiramate exhibited dose-dependent inhibition of action potential firing evoked by current injections into CA1 pyramidal neurons of KA-SE rats. In subiculum, KA-SE rats had frequent interictal spikes associated with high frequency oscillations while only rare spontaneous EPSPs were recorded in saline controls. Our experiments revealed that the hippocampal formation of aged epileptic rats shares features of hyperexcitability previously described in young adult epileptic rats using the KA model.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Neurology, University of Texas Health Science Center at San Antonio, TX, 78229, USA
- Departments of Surgery and Neuroscience & Experimental Therapeutics, Texas A&M Health Science Center, Temple, TX 76504, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Gleb P. Tolstykh
- Department of Neurology, University of Texas Health Science Center at San Antonio, TX, 78229, USA
| | - Russell M. Sanchez
- Departments of Surgery and Neuroscience & Experimental Therapeutics, Texas A&M Health Science Center, Temple, TX 76504, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Jose E. Cavazos
- Department of Neurology, University of Texas Health Science Center at San Antonio, TX, 78229, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, TX, 78229, USA
- Department of Physiology, University of Texas Health Science Center at San Antonio, TX, 78229, USA
- San Antonio VA Epilepsy Center of Excellence, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| |
Collapse
|
33
|
Arisi GM, Ruch M, Foresti ML, Mukherjee S, Ribak CE, Shapiro LA. Astrocyte Alterations in the Hippocampus Following Pilocarpine-induced Seizures in Aged Rats. Aging Dis 2011; 2:294-300. [PMID: 22396881 PMCID: PMC3295071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 08/25/2011] [Accepted: 08/26/2011] [Indexed: 05/31/2023] Open
Abstract
It is known that the incidence of epilepsy increases with age, but only a few studies have investigated the consequences and mechanisms of seizure and epilepsy in aged animals. Astrocytic changes are known to directly influence neuronal excitability and seizure susceptibility. However, information regarding alterations to astrocytes after seizures in aged animals is lacking in the literature. In the present study, the density and morphology of astrocytes expressing GFAP were investigated in the hippocampus of aged rats that experienced status epilepticus induced by pilocarpine. One month after seizures, astrocytes in aged rats have increased volume and present activated morphology. Despite these morphological changes, the density of astrocytes was not altered in the hippocampus of aged rats after seizures.
Collapse
Affiliation(s)
- Gabriel M Arisi
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Megan Ruch
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Maira L Foresti
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Sanjib Mukherjee
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
| | - Charles E Ribak
- Department of Anatomy and Neurobiology, University of California at Irvine, School of Medicine, Irvine, CA 92697-1275, USA
| | - Lee A Shapiro
- Scott & White Hospital, Neuroscience Research Institute, Temple, TX, USA
- Central Texas Veterans Health System, Temple, TX, USA
- Department of Surgery, Neurosurgery and Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
| |
Collapse
|
34
|
Hattiangady B, Kuruba R, Shetty AK. Acute Seizures in Old Age Leads to a Greater Loss of CA1 Pyramidal Neurons, an Increased Propensity for Developing Chronic TLE and a Severe Cognitive Dysfunction. Aging Dis 2011; 2:1-17. [PMID: 21339903 PMCID: PMC3041587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 01/26/2010] [Accepted: 01/26/2010] [Indexed: 05/30/2023] Open
Abstract
The aged population displays an enhanced risk for developing acute seizure (AS) activity. However, it is unclear whether AS activity in old age would result in a greater magnitude of hippocampal neurodegeneration and inflammation, and an increased predilection for developing chronic temporal lobe epilepsy (TLE) and cognitive dysfunction. Therefore, we addressed these issues in young-adult (5-months old) and aged (22-months old) F344 rats after three-hours of AS activity, induced through graded intraperitoneal injections of kainic acid (KA), and terminated through a diazepam injection. During the three-hours of AS activity, both young adult and aged groups exhibited similar numbers of stage-V motor seizures but the numbers of stage-IV motor seizures were greater in the aged group. In both age groups, three-hour AS activity induced degeneration of 50-55% of neurons in the dentate hilus, 22-32% of neurons in the granule cell layer and 49-52% neurons in the CA3 pyramidal cell layer without showing any interaction between the age and AS activity. However, degeneration of neurons in the CA1 pyramidal cell layer showed a clear interaction between the age and AS activity (12% in the young adult group and 56% in the aged group), suggesting that an advanced age makes the CA1 pyramidal neurons more susceptible to die with AS activity. The extent of inflammation measured through the numbers of activated microglial cells was similar between the two age groups. Interestingly, the predisposition for developing chronic TLE at 2-3 months after AS activity was 60% for young adult rats but 100% for aged rats. Moreover, both frequency & intensity of spontaneous recurrent seizures in the chronic phase after AS activity were 6-12 folds greater in aged rats than in young adult rats. Furthermore, aged rats lost their ability for spatial learning even in a scrupulous eleven-session water maze learning paradigm after AS activity, in divergence from young adult rats which retained the ability for spatial learning but had memory retrieval dysfunction after AS activity. Thus, AS activity in old age results in a greater loss of hippocampal CA1 pyramidal neurons, an increased propensity for developing robust chronic TLE, and a severe cognitive dysfunction.
Collapse
Affiliation(s)
| | | | - Ashok K. Shetty
- Correspondence should be addressed to: Ashok K. Shetty, M.Sc., Ph.D., Division of Neurosurgery, DUMC Box 3807, Duke University Medical Center, Durham NC 27710. E-mail:
| |
Collapse
|
35
|
Shetty AK, Hattiangady B, Rao MS. Vulnerability of hippocampal GABA-ergic interneurons to kainate-induced excitotoxic injury during old age. J Cell Mol Med 2010. [PMID: 20141618 DOI: 10.1111/j.1582-4934.2008.00675.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Hippocampal inhibitory interneurons expressing glutamate decarboxylase-67 (GAD-67) considerably decline in number during old age. Studies in young adult animals further suggest that hippocampal GAD-67+ interneuron population is highly vulnerable to excitotoxic injury. However, the relative susceptibility of residual GAD-67+ interneurons in the aged hippocampus to excitotoxic injury is unknown. To elucidate this, using both adult and aged F344 rats, we performed stereological counting of GAD-67+ interneurons in different layers of the dentate gyrus and CA1 & CA3 sub-fields, at 3 months post-excitotoxic hippocampal injury inflicted through an intracerebroventricular administration of kainic acid (KA). Substantial reductions of GAD-67+ interneurons were found in all hippocampal layers and sub-fields after KA-induced injury in adult animals. Contrastingly, there was no significant change in GAD-67+ interneuron population in any of the hippocampal layers and sub-fields following similar injury in aged animals. Furthermore, the stability of GAD-67+ interneurons in aged rats after KA was not attributable to milder injury, as the overall extent of KA-induced hippocampal principal neuron loss was comparable between adult and aged rats. Interestingly, because of the age-related disparity in vulnerability of interneurons to injury, the surviving GAD-67+ interneuron population in the injured aged hippocampus remained comparable to that observed in the injured adult hippocampus despite enduring significant reductions in interneuron number with aging. Thus, unlike in the adult hippocampus, an excitotoxic injury to the aged hippocampus does not result in significantly decreased numbers of GAD-67+ interneurons. Persistence of GAD-67+ interneuron population in the injured aged hippocampus likely reflects an age-related change in the response of GAD-67+ interneurons to excitotoxic hippocampal injury. These results have implications towards understanding mechanisms underlying the evolution of initial precipitating injury into temporal lobe epilepsy in the elderly population.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC, USA.
| | | | | |
Collapse
|
36
|
Shetty AK, Hattiangady B, Rao MS. Vulnerability of hippocampal GABA-ergic interneurons to kainate-induced excitotoxic injury during old age. J Cell Mol Med 2010; 13:2408-23. [PMID: 20141618 DOI: 10.1111/j.1582-4934.2009.00675.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Hippocampal inhibitory interneurons expressing glutamate decarboxylase-67 (GAD-67) considerably decline in number during old age. Studies in young adult animals further suggest that hippocampal GAD-67+ interneuron population is highly vulnerable to excitotoxic injury. However, the relative susceptibility of residual GAD-67+ interneurons in the aged hippocampus to excitotoxic injury is unknown. To elucidate this, using both adult and aged F344 rats, we performed stereological counting of GAD-67+ interneurons in different layers of the dentate gyrus and CA1 & CA3 sub-fields, at 3 months post-excitotoxic hippocampal injury inflicted through an intracerebroventricular administration of kainic acid (KA). Substantial reductions of GAD-67+ interneurons were found in all hippocampal layers and sub-fields after KA-induced injury in adult animals. Contrastingly, there was no significant change in GAD-67+ interneuron population in any of the hippocampal layers and sub-fields following similar injury in aged animals. Furthermore, the stability of GAD-67+ interneurons in aged rats after KA was not attributable to milder injury, as the overall extent of KA-induced hippocampal principal neuron loss was comparable between adult and aged rats. Interestingly, because of the age-related disparity in vulnerability of interneurons to injury, the surviving GAD-67+ interneuron population in the injured aged hippocampus remained comparable to that observed in the injured adult hippocampus despite enduring significant reductions in interneuron number with aging. Thus, unlike in the adult hippocampus, an excitotoxic injury to the aged hippocampus does not result in significantly decreased numbers of GAD-67+ interneurons. Persistence of GAD-67+ interneuron population in the injured aged hippocampus likely reflects an age-related change in the response of GAD-67+ interneurons to excitotoxic hippocampal injury. These results have implications towards understanding mechanisms underlying the evolution of initial precipitating injury into temporal lobe epilepsy in the elderly population.
Collapse
Affiliation(s)
- Ashok K Shetty
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC, USA.
| | | | | |
Collapse
|
37
|
Abstract
Aged animals have been used by researchers to better understand the differences between the young and the aged brain and how these differences may provide insight into the mechanisms of acute seizures and epilepsy in the elderly. To date, there have been relatively few studies dedicated to the modeling of acute seizures and epilepsy in aged, healthy animals. Inherent challenges to this area of research include the costs associated with the purchase and maintenance of older animals and, at times, the unexpected and potentially confounding comorbidities associated with aging. However, recent studies using a variety of in vivo and in vitro models of acute seizures and epilepsy in mice and rats have built upon early investigations in the field, all of which has provided an expanded vision of seizure generation and epileptogenesis in the aged brain. Results of these studies could potentially translate to new and tailored interventional approaches that limit or prevent the development of epilepsy in the elderly.
Collapse
Affiliation(s)
- Kevin M Kelly
- Drexel University College of Medicine, Center for Neuroscience Research, Allegheny-Singer Research Institute, Allegheny General Hospital Pittsburgh, Pittsburgh, PA 15212-4772, USA.
| |
Collapse
|
38
|
Pankratova S, Kiryushko D, Sonn K, Soroka V, Køhler LB, Rathje M, Gu B, Gotfryd K, Clausen O, Zharkovsky A, Bock E, Berezin V. Neuroprotective properties of a novel, non-haematopoietic agonist of the erythropoietin receptor. Brain 2010; 133:2281-94. [DOI: 10.1093/brain/awq101] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
39
|
Schauwecker PE. Neuroprotection by glutamate receptor antagonists against seizure-induced excitotoxic cell death in the aging brain. Exp Neurol 2010; 224:207-18. [PMID: 20353782 DOI: 10.1016/j.expneurol.2010.03.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Revised: 03/17/2010] [Accepted: 03/21/2010] [Indexed: 10/19/2022]
Abstract
We previously have identified phenotypic differences in susceptibility to hippocampal seizure-induced cell death among two inbred strains of mice. We have also reported that the age-related increased susceptibility to the neurotoxic effects of seizure-induced injury is regulated in a strain-dependent manner. In the present study, we wanted to begin to determine the pharmacological mechanism that contributes to variability in the response to the neurotoxic effects of kainate. Thus, we compared the effects of the NMDA receptor antagonist, MK-801 and of the AMPA receptor antagonist NBQX on hippocampal damage in the kainate model of seizure-induced excitotoxic cell death in young, middle-aged, and aged C57BL/6 and FVB/N mice, when given 90 min following kainate-induced status epilepticus. Following kainate injections, mice were scored for seizure activity and brains from mice in each age and antagonist group were processed for light microscopic histopathologic evaluation 7 days following kainate administration to evaluate the severity of seizure-induced injury. Administration of MK-801 significantly reduced the extent of hippocampal damage in young, mature and aged FVB/N mice, while application of NBQX was only effective at attenuating cell death in young and aged mice throughout all hippocampal subfields. Our results suggest that both NMDA and non-NMDA receptors are involved in kainate-induced cell death in the mouse and suggest that aging may differentially affect the ability of neuroprotectants to protect against hippocampal damage. Differences in the effectiveness of these two antagonists could result from differential regulation of glutamatergic neurotransmitter systems or ion channel specificity.
Collapse
Affiliation(s)
- P Elyse Schauwecker
- Department of Cell and Neurobiology, USC Keck School of Medicine, 1333 San Pablo Street, BMT 403, Los Angeles, CA 90089-9112, USA.
| |
Collapse
|
40
|
Stewart I. Environmental risk factors for temporal lobe epilepsy – Is prenatal exposure to the marine algal neurotoxin domoic acid a potentially preventable cause? Med Hypotheses 2010; 74:466-81. [DOI: 10.1016/j.mehy.2009.10.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 10/04/2009] [Indexed: 01/29/2023]
|
41
|
Kienzler F, Norwood BA, Sloviter RS. Hippocampal injury, atrophy, synaptic reorganization, and epileptogenesis after perforant pathway stimulation-induced status epilepticus in the mouse. J Comp Neurol 2009; 515:181-96. [PMID: 19412934 DOI: 10.1002/cne.22059] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Prolonged dentate granule cell discharges produce hippocampal injury and chronic epilepsy in rats. In preparing to study this epileptogenic process in genetically altered mice, we determined whether the background strain used to generate most genetically altered mice, the C57BL/6 mouse, is vulnerable to stimulation-induced seizure-induced injury. This was necessary because C57BL/6 mice are reportedly resistant to the neurotoxic effects of kainate-induced seizures, which we hypothesized to be related to strain differences in kainate's effects, rather than genetic differences in intrinsic neuronal vulnerability. Bilateral perforant pathway stimulation-induced granule cell discharge for 4 hours under urethane anesthesia produced degeneration of glutamate receptor subunit 2 (GluR2)-positive hilar mossy cells and peptide-containing interneurons in both FVB/N (kainate-vulnerable) and C57BL/6 (kainate-resistant) mice, indicating no strain differences in neuronal vulnerability to seizure activity. Granule cell discharge for 2 hours in C57BL/6 mice destroyed most GluR2-positive dentate hilar mossy cells, but not peptide-containing hilar interneurons, indicating that mossy cells are the neurons most vulnerable to this insult. Stimulation for 24 hours caused extensive hippocampal neuron loss and injury to the septum and entorhinal cortex, but no other detectable damage. Mice stimulated for 24 hours developed hippocampal sclerosis, granule cell mossy fiber sprouting, and chronic epilepsy, but not the granule cell layer hypertrophy (granule cell dispersion) produced by intrahippocampal kainate. These results demonstrate that perforant pathway stimulation in mice reliably reproduces the defining features of human mesial temporal lobe epilepsy with hippocampal sclerosis. Experimental studies in transgenic or knockout mice are feasible if electrical stimulation is used to produce controlled epileptogenic insults.
Collapse
Affiliation(s)
- Friederike Kienzler
- Departments of Pharmacology and Neurology, University of Arizona College of Medicine, Tucson, Arizona 85724, USA
| | | | | |
Collapse
|