1
|
Leem E, Kim S, Sharma C, Nam Y, Kim TY, Shin M, Lee SG, Kim J, Kim SR. Inhibition of Granule Cell Dispersion and Seizure Development by Astrocyte Elevated Gene-1 in a Mouse Model of Temporal Lobe Epilepsy. Biomolecules 2024; 14:380. [PMID: 38540798 PMCID: PMC10968595 DOI: 10.3390/biom14030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Although granule cell dispersion (GCD) in the hippocampus is known to be an important feature associated with epileptic seizures in temporal lobe epilepsy (TLE), the endogenous molecules that regulate GCD are largely unknown. In the present study, we have examined whether there is any change in AEG-1 expression in the hippocampus of a kainic acid (KA)-induced mouse model of TLE. In addition, we have investigated whether the modulation of astrocyte elevated gene-1 (AEG-1) expression in the dentate gyrus (DG) by intracranial injection of adeno-associated virus 1 (AAV1) influences pathological phenotypes such as GCD formation and seizure susceptibility in a KA-treated mouse. We have identified that the protein expression of AEG-1 is upregulated in the DG of a KA-induced mouse model of TLE. We further demonstrated that AEG-1 upregulation by AAV1 delivery in the DG-induced anticonvulsant activities such as the delay of seizure onset and inhibition of spontaneous recurrent seizures (SRS) through GCD suppression in the mouse model of TLE, while the inhibition of AEG-1 expression increased susceptibility to seizures. The present observations suggest that AEG-1 is a potent regulator of GCD formation and seizure development associated with TLE, and the significant induction of AEG-1 in the DG may have therapeutic potential against epilepsy.
Collapse
Affiliation(s)
- Eunju Leem
- BK21 FOUR KNU Creative BioResearch Group, School of Life Science, Kyungpook National University, Daegu 41566, Republic of Korea; (E.L.); (S.K.); (C.S.); (T.Y.K.)
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
- Efficacy Evaluation Department, New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Sehwan Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Science, Kyungpook National University, Daegu 41566, Republic of Korea; (E.L.); (S.K.); (C.S.); (T.Y.K.)
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Chanchal Sharma
- BK21 FOUR KNU Creative BioResearch Group, School of Life Science, Kyungpook National University, Daegu 41566, Republic of Korea; (E.L.); (S.K.); (C.S.); (T.Y.K.)
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Tae Yeon Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Science, Kyungpook National University, Daegu 41566, Republic of Korea; (E.L.); (S.K.); (C.S.); (T.Y.K.)
| | - Minsang Shin
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Seok-Geun Lee
- Department of Biomedical Science & Technology and BioNanocomposite Research Center, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Sang Ryong Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Science, Kyungpook National University, Daegu 41566, Republic of Korea; (E.L.); (S.K.); (C.S.); (T.Y.K.)
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea;
| |
Collapse
|
2
|
AlRuwaili R, Al-Kuraishy HM, Al-Gareeb AI, Ali NH, Alexiou A, Papadakis M, Saad HM, Batiha GES. The Possible Role of Brain-derived Neurotrophic Factor in Epilepsy. Neurochem Res 2024; 49:533-547. [PMID: 38006577 PMCID: PMC10884085 DOI: 10.1007/s11064-023-04064-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Epilepsy is a neurological disease characterized by repeated seizures. Despite of that the brain-derived neurotrophic factor (BDNF) is implicated in the pathogenesis of epileptogenesis and epilepsy, BDNF may have a neuroprotective effect against epilepsy. Thus, the goal of the present review was to highlight the protective and detrimental roles of BDNF in epilepsy. In this review, we also try to find the relation of BDNF with other signaling pathways and cellular processes including autophagy, mTOR pathway, progranulin (PGN), and α-Synuclein (α-Syn) which negatively and positively regulate BDNF/tyrosine kinase receptor B (TrkB) signaling pathway. Therefore, the assessment of BDNF levels in epilepsy should be related to other neuronal signaling pathways and types of epilepsy in both preclinical and clinical studies. In conclusion, there is a strong controversy concerning the potential role of BDNF in epilepsy. Therefore, preclinical, molecular, and clinical studies are warranted in this regard.
Collapse
Affiliation(s)
- Raed AlRuwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, Wien, 1030, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
3
|
Sri Hari A, Banerji R, Liang LP, Fulton RE, Huynh CQ, Fabisiak T, McElroy PB, Roede JR, Patel M. Increasing glutathione levels by a novel posttranslational mechanism inhibits neuronal hyperexcitability. Redox Biol 2023; 67:102895. [PMID: 37769522 PMCID: PMC10539966 DOI: 10.1016/j.redox.2023.102895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023] Open
Abstract
Glutathione (GSH) depletion, and impaired redox homeostasis have been observed in experimental animal models and patients with epilepsy. Pleiotropic strategies that elevate GSH levels via transcriptional regulation have been shown to significantly decrease oxidative stress and seizure frequency, increase seizure threshold, and rescue certain cognitive deficits. Whether elevation of GSH per se alters neuronal hyperexcitability remains unanswered. We previously showed that thiols such as dimercaprol (DMP) elevate GSH via post-translational activation of glutamate cysteine ligase (GCL), the rate limiting GSH biosynthetic enzyme. Here, we asked if elevation of cellular GSH by DMP altered neuronal hyperexcitability in-vitro and in-vivo. Treatment of primary neuronal-glial cerebrocortical cultures with DMP elevated GSH and inhibited a voltage-gated potassium channel blocker (4-aminopyridine, 4AP) induced neuronal hyperexcitability. DMP increased GSH in wildtype (WT) zebrafish larvae and significantly attenuated convulsant pentylenetetrazol (PTZ)-induced acute 'seizure-like' swim behavior. DMP treatment increased GSH and inhibited convulsive, spontaneous 'seizure-like' swim behavior in the Dravet Syndrome (DS) zebrafish larvae (scn1Lab). Furthermore, DMP treatment significantly decreased spontaneous electrographic seizures and associated seizure parameters in scn1Lab zebrafish larvae. We investigated the role of the redox-sensitive mammalian target of rapamycin (mTOR) pathway due to the presence of several cysteine-rich proteins and their involvement in regulating neuronal excitability. Treatment of primary neuronal-glial cerebrocortical cultures with 4AP or l-buthionine-(S,R)-sulfoximine (BSO), an irreversible inhibitor of GSH biosynthesis, significantly increased mTOR complex I (mTORC1) activity which was rescued by pre-treatment with DMP. Furthermore, BSO-mediated GSH depletion oxidatively modified the tuberous sclerosis protein complex (TSC) consisting of hamartin (TSC1), tuberin (TSC2), and TBC1 domain family member 7 (TBC1D7) which are critical negative regulators of mTORC1. In summary, our results suggest that DMP-mediated GSH elevation by a novel post-translational mechanism can inhibit neuronal hyperexcitability both in-vitro and in-vivo and a plausible link is the redox sensitive mTORC1 pathway.
Collapse
Affiliation(s)
- Ashwini Sri Hari
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rajeswari Banerji
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ruth E Fulton
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Christopher Quoc Huynh
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Timothy Fabisiak
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Pallavi Bhuyan McElroy
- The Janssen Pharmaceutical Companies of Johnson & Johnson, Greater Philadelphia Area, Horsham, PA, 19044, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Zhao W, Xie C, Zhang X, Liu J, Liu J, Xia Z. Advances in the mTOR signaling pathway and its inhibitor rapamycin in epilepsy. Brain Behav 2023; 13:e2995. [PMID: 37221133 PMCID: PMC10275542 DOI: 10.1002/brb3.2995] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION Epilepsy is one of the most common and serious brain syndromes and has adverse consequences on a patient's neurobiological, cognitive, psychological, and social wellbeing, thereby threatening their quality of life. Some patients with epilepsy experience poor treatment effects due to the unclear pathophysiological mechanisms of the syndrome. Dysregulation of the mammalian target of the rapamycin (mTOR) pathway is thought to play an important role in the onset and progression of some epilepsies. METHODS This review summarizes the role of the mTOR signaling pathway in the pathogenesis of epilepsy and the prospects for the use of mTOR inhibitors. RESULTS The mTOR pathway functions as a vital mediator in epilepsy development through diverse mechanisms, indicating that the it has great potential as an effective target for epilepsy therapy. The excessive activation of mTOR signaling pathway leads to structural changes in neurons, inhibits autophagy, exacerbates neuron damage, affects mossy fiber sprouting, enhances neuronal excitability, increases neuroinflammation, and is closely associated with tau upregulation in epilepsy. A growing number of studies have demonstrated that mTOR inhibitors exhibit significant antiepileptic effects in both clinical applications and animal models. Specifically, rapamycin, a specific inhibitor of TOR, reduces the intensity and frequency of seizures. Clinical studies in patients with tuberous sclerosis complex have shown that rapamycin has the function of reducing seizures and improving this disease. Everolimus, a chemically modified derivative of rapamycin, has been approved as an added treatment to other antiepileptic medicines. Further explorations are needed to evaluate the therapeutic efficacy and application value of mTOR inhibitors in epilepsy. CONCLUSIONS Targeting the mTOR signaling pathway provides a promising prospect for the treatment of epilepsy.
Collapse
Affiliation(s)
- Wei Zhao
- Department of GerontologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Cong Xie
- Department of GerontologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Xu Zhang
- Department of GerontologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Ju Liu
- Laboratory of Microvascular MedicineMedical Research CenterShandong Provincial Qianfoshan Hospital, Shandong UniversityJinanChina
| | - Jinzhi Liu
- Department of GerontologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Department of NeurologyLiaocheng People's Hospital and Liaocheng Clinical School of Shandong First Medical UniversityLiaochengChina
- Department of GerontologyCheeloo College of MedicineShandong Provincial Qianfoshan Hospital, Shandong UniversityJinanChina
- Department of Geriatric NeurologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Zhangyong Xia
- Department of NeurologyLiaocheng People's Hospital and Liaocheng Clinical School of Shandong First Medical UniversityLiaochengChina
- Department of NeurologyCheeloo College of MedicineLiaocheng People's Hospital, Shandong UniversityJinanChina
| |
Collapse
|
5
|
Kasahara Y, Nakashima H, Nakashima K. Seizure-induced hilar ectopic granule cells in the adult dentate gyrus. Front Neurosci 2023; 17:1150283. [PMID: 36937666 PMCID: PMC10017466 DOI: 10.3389/fnins.2023.1150283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Epilepsy is a chronic neurological disorder characterized by hypersynchronous spontaneous recurrent seizures, and affects approximately 50 million people worldwide. Cumulative evidence has revealed that epileptogenic insult temporarily increases neurogenesis in the hippocampus; however, a fraction of the newly generated neurons are integrated abnormally into the existing neural circuits. The abnormal neurogenesis, including ectopic localization of newborn neurons in the hilus, formation of abnormal basal dendrites, and disorganization of the apical dendrites, rewires hippocampal neural networks and leads to the development of spontaneous seizures. The central roles of hilar ectopic granule cells in regulating hippocampal excitability have been suggested. In this review, we introduce recent findings about the migration of newborn granule cells to the dentate hilus after seizures and the roles of seizure-induced ectopic granule cells in the epileptic brain. In addition, we delineate possible intrinsic and extrinsic mechanisms underlying this abnormality. Finally, we suggest that the regulation of seizure-induced ectopic cells can be a promising target for epilepsy therapy and provide perspectives on future research directions.
Collapse
|
6
|
Cutia CA, Leverton LK, Ge X, Youssef R, Raetzman LT, Christian-Hinman CA. Phenotypic differences based on lateralization of intrahippocampal kainic acid injection in female mice. Exp Neurol 2022; 355:114118. [PMID: 35597270 PMCID: PMC10462257 DOI: 10.1016/j.expneurol.2022.114118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/17/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022]
Abstract
Clinical evidence indicates that patients with temporal lobe epilepsy (TLE) often show differential outcomes of comorbid conditions in relation to the lateralization of the seizure focus. A particularly strong relationship exists between the side of seizure focus and the propensity for distinct reproductive endocrine comorbidities in women with TLE. Therefore, here we evaluated whether targeting of left or right dorsal hippocampus for intrahippocampal kainic acid (IHKA) injection, a model of TLE, produces different outcomes in hippocampal granule cell dispersion, body weight gain, and multiple measures of reproductive endocrine dysfunction in female mice. One, two, and four months after IHKA or saline injection, in vivo measurements of estrous cycles and weight were followed by ex vivo examination of hippocampal dentate granule cell dispersion, circulating ovarian hormone and corticosterone levels, ovarian morphology, and pituitary gene expression. IHKA mice with right-targeted injection (IHKA-R) showed greater granule cell dispersion and pituitary Fshb expression compared to mice with left-targeted injection (IHKA-L). By contrast, pituitary expression of Lhb and Gnrhr were higher in IHKA-L mice compared to IHKA-R, but these values were not different from respective saline-injected controls. IHKA-L mice also showed an increased rate of weight gain compared to IHKA-R mice. Increases in estrous cycle length, however, were similar in both IHKA-L and IHKA-R mice. These findings indicate that although major reproductive endocrine dysfunction phenotypes present similarly after targeting left or right dorsal hippocampus for IHKA injection, distinct underlying mechanisms based on lateralization of epileptogenic insult may contribute to produce similar emergent reproductive endocrine outcomes.
Collapse
Affiliation(s)
- Cathryn A Cutia
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Leanna K Leverton
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiyu Ge
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Rana Youssef
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Lori T Raetzman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, 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
|
7
|
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
|
8
|
Overexpression of Lin28A in neural progenitor cells in vivo does not lead to brain tumor formation but results in reduced spine density. Acta Neuropathol Commun 2021; 9:185. [PMID: 34801069 PMCID: PMC8606090 DOI: 10.1186/s40478-021-01289-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/02/2021] [Indexed: 11/10/2022] Open
Abstract
LIN28A overexpression has been identified in malignant brain tumors called embryonal tumors with multilayered rosettes (ETMR) but its specific role during brain development remains largely unknown. Radial glia cells of the ventricular zone (VZ) are proposed as a cell of origin for ETMR. We asked whether an overexpression of LIN28A in such cells might affect brain development or result in the formation of brain tumors.Constitutive overexpression of LIN28A in hGFAP-cre::lsl-Lin28A (GL) mice led to a transient increase of proliferation in the cortical VZ at embryonic stages but no postnatal brain tumor formation. Postnatally, GL mice displayed a pyramidal cell layer dispersion of the hippocampus and altered spine and dendrite morphology, including reduced dendritic spine densities in the hippocampus and cortex. GL mice displayed hyperkinetic activity and differential quantitative MS-based proteomics revealed altered time dependent molecular functions regarding mRNA processing and spine morphogenesis. Phosphoproteomic analyses indicated a downregulation of mTOR pathway modulated proteins such as Map1b being involved in microtubule dynamics.In conclusion, we show that Lin28A overexpression transiently increases proliferation of neural precursor cells but it is not sufficient to drive brain tumors in vivo. In contrast, Lin28A impacts on protein abundancy patterns related to spine morphogenesis and phosphorylation levels of proteins involved in microtubule dynamics, resulting in decreased spine densities of neurons in the hippocampus and cortex as well as in altered behavior. Our work provides new insights into the role of LIN28A for neuronal morphogenesis and development and may reveal future targets for treatment of ETMR patients.
Collapse
|
9
|
Ma KG, Hu HB, Zhou JS, Ji C, Yan QS, Peng SM, Ren LD, Yang BN, Xiao XL, Ma YB, Wu F, Si KW, Wu XL, Liu JX. Neuronal Glypican4 promotes mossy fiber sprouting through the mTOR pathway after pilocarpine-induced status epilepticus in mice. Exp Neurol 2021; 347:113918. [PMID: 34748756 DOI: 10.1016/j.expneurol.2021.113918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 02/07/2023]
Abstract
In temporal lobe epilepsy (TLE), abnormal axon guidance and synapse formation lead to sprouting of mossy fibers in the hippocampus, which is one of the most consistent pathological findings in patients and animal models with TLE. Glypican 4 (Gpc4) belongs to the heparan sulfate proteoglycan family, which play an important role in axon guidance and excitatory synapse formation. However, the role of Gpc4 in the development of mossy fibers sprouting (MFS) and its underlying mechanism remain unknown. Using a pilocarpine-induced mice model of epilepsy, we showed that Gpc4 expression was significantly increased in the stratum granulosum of the dentate gyrus at 1 week after status epilepticus (SE). Using Gpc4 overexpression or Gpc4 shRNA lentivirus to regulate the Gpc4 level in the dentate gyrus, increased or decreased levels of netrin-1, SynI, PSD-95, and Timm score were observed in the dentate gyrus, indicating a crucial role of Gpc4 in modulating the development of functional MFS. The observed effects of Gpc4 on MFS were significantly antagonized when mice were treated with L-leucine or rapamycin, an agonist or antagonist of the mammalian target of rapamycin (mTOR) signal, respectively, demonstrating that mTOR pathway is an essential requirement for Gpc4-regulated MFS. Additionally, the attenuated spontaneous recurrent seizures (SRSs) were observed during chronic stage of the disease by suppressing the Gpc4 expression after SE. Altogether, our findings demonstrate a novel control of neuronal Gpc4 on the development of MFS through the mTOR pathway after pilocarpine-induced SE. Our results also strongly suggest that Gpc4 may serve as a promising target for antiepileptic studies.
Collapse
Affiliation(s)
- Kai-Ge Ma
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Hai-Bo Hu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Jin-Song Zhou
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Chao Ji
- Qide College, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Qi-Sheng Yan
- Qide College, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Si-Ming Peng
- Zonglian College, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Lan-Dong Ren
- Zonglian College, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Bing-Nan Yang
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Xin-Li Xiao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Yan-Bing Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Feng Wu
- Center of Teaching and Experiment for Medical Post Graduates, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Kai-Wei Si
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China
| | - Xiao-Lin Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China.
| | - Jian-Xin Liu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, China.
| |
Collapse
|
10
|
Szala-Rycaj J, Zagaja M, Szewczyk A, Andres-Mach M. Selected flavonoids and their role in the treatment of epilepsy – a review of the latest reports from experimental studies. Acta Neurobiol Exp (Wars) 2021. [DOI: 10.21307/ane-2021-014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
11
|
Welzel L, Bergin DH, Schidlitzki A, Twele F, Johne M, Klein P, Löscher W. Systematic evaluation of rationally chosen multitargeted drug combinations: a combination of low doses of levetiracetam, atorvastatin and ceftriaxone exerts antiepileptogenic effects in a mouse model of acquired epilepsy. Neurobiol Dis 2020; 149:105227. [PMID: 33347976 DOI: 10.1016/j.nbd.2020.105227] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/24/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023] Open
Abstract
Epileptogenesis, the gradual process that leads to epilepsy after brain injury or genetic mutations, is a complex network phenomenon, involving a variety of morphological, biochemical and functional brain alterations. Although risk factors for developing epilepsy are known, there is currently no treatment available to prevent epilepsy. We recently proposed a multitargeted, network-based approach to prevent epileptogenesis by rationally combining clinically available drugs and provided first proof-of-concept that this strategy is effective. Here we evaluated eight novel rationally chosen combinations of 14 drugs with mechanisms that target different epileptogenic processes. The combinations consisted of 2-4 different drugs per combination and were administered systemically over 5 days during the latent epileptogenic period in the intrahippocampal kainate mouse model of acquired temporal lobe epilepsy, starting 6 h after kainate. Doses and dosing intervals were based on previous pharmacokinetic and tolerability studies in mice. The incidence and frequency of spontaneous electrographic and electroclinical seizures were recorded by continuous (24/7) video linked EEG monitoring done for seven days at 4 and 12 weeks post-kainate, i.e., long after termination of drug treatment. Compared to vehicle controls, the most effective drug combination consisted of low doses of levetiracetam, atorvastatin and ceftriaxone, which markedly reduced the incidence of electrographic seizures (by 60%; p<0.05) and electroclinical seizures (by 100%; p<0.05) recorded at 12 weeks after kainate. This effect was lost when higher doses of the three drugs were administered, indicating a synergistic drug-drug interaction at the low doses. The potential mechanisms underlying this interaction are discussed. We have discovered a promising novel multitargeted combination treatment for modifying the development of acquired epilepsy.
Collapse
Affiliation(s)
- Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - David H Bergin
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
12
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
13
|
Novel brain permeant mTORC1/2 inhibitors are as efficacious as rapamycin or everolimus in mouse models of acquired partial epilepsy and tuberous sclerosis complex. Neuropharmacology 2020; 180:108297. [PMID: 32890589 DOI: 10.1016/j.neuropharm.2020.108297] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/24/2022]
Abstract
Mechanistic target of rapamycin (mTOR) regulates cell proliferation, growth and survival, and is activated in cancer and neurological disorders, including epilepsy. The rapamycin derivative ("rapalog") everolimus, which allosterically inhibits the mTOR pathway, is approved for the treatment of partial epilepsy with spontaneous recurrent seizures (SRS) in individuals with tuberous sclerosis complex (TSC). In contrast to the efficacy in TSC, the efficacy of rapalogs on SRS in other types of epilepsy is equivocal. Furthermore, rapalogs only poorly penetrate into the brain and are associated with peripheral adverse effects, which may compromise their therapeutic efficacy. Here we compare the antiseizure efficacy of two novel, brain-permeable ATP-competitive and selective mTORC1/2 inhibitors, PQR620 and PQR626, and the selective dual pan-PI3K/mTORC1/2 inhibitor PQR530 in two mouse models of chronic epilepsy with SRS, the intrahippocampal kainate (IHK) mouse model of acquired temporal lobe epilepsy and Tsc1GFAP CKO mice, a well-characterized mouse model of epilepsy in TSC. During prolonged treatment of IHK mice with rapamycin, everolimus, PQR620, PQR626, or PQR530; only PQR620 exerted a transient antiseizure effect on SRS, at well tolerated doses whereas the other compounds were ineffective. In contrast, all of the examined compounds markedly suppressed SRS in Tsc1GFAP CKO mice during chronic treatment at well tolerated doses. Thus, against our expectation, no clear differences in antiseizure efficacy were found across the three classes of mTOR inhibitors examined in mouse models of genetic and acquired epilepsies. The main advantage of the novel 1,3,5-triazine derivatives is their excellent tolerability compared to rapalogs, which would favor their development as new therapies for TORopathies such as TSC.
Collapse
|
14
|
Jeong KH, Cho KO, Lee MY, Kim SY, Kim WJ. Vascular endothelial growth factor receptor-3 regulates astroglial glutamate transporter-1 expression via mTOR activation in reactive astrocytes following pilocarpine-induced status epilepticus. Glia 2020; 69:296-309. [PMID: 32835451 DOI: 10.1002/glia.23897] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022]
Abstract
Recent evidence has shown that the vascular endothelial growth factor (VEGF) system plays a crucial role in several neuropathological processes. We previously reported an upregulation of VEGF-C and its receptor, VEGFR-3, in reactive astrocytes after the onset of status epilepticus (SE). However, it remains unknown, which molecules act as downstream signals following VEGFR-3 upregulation, and are involved in reactive astrogliosis after SE. Therefore, we investigated whether VEGFR-3 upregulation within reactive astrocytes is associated with the activation of mammalian target of rapamycin (mTOR) signaling, which we confirmed by assaying for the phosphorylated form of S6 protein (pS6), and whether VEGFR-3-mediated mTOR activation induces astroglial glutamate transporter-1 (GLT-1) expression in the hippocampus after pilocarpine-induced SE. We found that spatiotemporal expression of pS6 was consistent with VEGFR-3 expression in the hippocampus after SE, and that both pS6 and VEGFR-3 were highly expressed in SE-induced reactive astrocytes. Treatment with the mTOR inhibitor rapamycin decreased astroglial VEGFR-3 expression and GLT-1 expression after SE. Treatment with a selective inhibitor for VEGFR-3 attenuated astroglial pS6 expression as well as suppressed GLT-1 expression and astroglial reactivity in the hippocampus after SE. These findings demonstrate that VEGFR-3-mediated mTOR activation could contribute to the regulation of GLT-1 expression in reactive astrocytes during the subacute phase of epilepsy. In conclusion, the present study suggests that VEGFR-3 upregulation in reactive astrocytes may play a role in preventing hyperexcitability induced by continued seizure activity.
Collapse
Affiliation(s)
- Kyoung Hoon Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mun-Yong Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong Yun Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Won-Joo Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Tanioka M, Park WK, Shim I, Kim K, Choi S, Kim UJ, Lee KH, Hong SK, Lee BH. Neuroprotection from Excitotoxic Injury by Local Administration of Lipid Emulsion into the Brain of Rats. Int J Mol Sci 2020; 21:ijms21082706. [PMID: 32295117 PMCID: PMC7215821 DOI: 10.3390/ijms21082706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 01/10/2023] Open
Abstract
Lipid emulsion was recently shown to attenuate cell death caused by excitotoxic conditions in the heart. There are key similarities between neurons and cardiomyocytes, such as excitability and conductibility, which yield vulnerability to excitotoxic conditions. However, systematic investigations on the protective effects of lipid emulsion in the central nervous system are still lacking. This study aimed to determine the neuroprotective effects of lipid emulsion in an in vivo rat model of kainic acid-induced excitotoxicity through intrahippocampal microinjections. Kainic acid and/or lipid emulsion-injected rats were subjected to the passive avoidance test and elevated plus maze for behavioral assessment. Rats were sacrificed at 24 h and 72 h after kainic acid injections for molecular study, including immunoblotting and qPCR. Brains were also cryosectioned for morphological analysis through cresyl violet staining and Fluorojade-C staining. Anxiety and memory functions were significantly preserved in 1% lipid emulsion-treated rats. Lipid emulsion was dose-dependent on the protein expression of β-catenin and the phosphorylation of GSK3-β and Akt. Wnt1 mRNA expression was elevated in lipid emulsion-treated rats compared to the vehicle. Neurodegeneration was significantly reduced mainly in the CA1 region with increased cell survival. Our results suggest that lipid emulsion has neuroprotective effects against excitotoxic conditions in the brain and may provide new insight into its potential therapeutic utility.
Collapse
Affiliation(s)
- Motomasa Tanioka
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (M.T.); (K.K.); (S.C.); (U.J.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Wyun Kon Park
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Insop Shim
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Kyeongmin Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (M.T.); (K.K.); (S.C.); (U.J.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Songyeon Choi
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (M.T.); (K.K.); (S.C.); (U.J.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Un Jeng Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (M.T.); (K.K.); (S.C.); (U.J.K.)
| | - Kyung Hee Lee
- Department of Dental Hygiene, Division of Health Science, Dongseo University, Busan 47011, Korea;
| | - Seong-Karp Hong
- Division of Biomedical Engineering, Mokwon University, Daejeon 35349, Korea;
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (M.T.); (K.K.); (S.C.); (U.J.K.)
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: + 82-2-2228-1711
| |
Collapse
|
16
|
Miziak B, Konarzewska A, Ułamek-Kozioł M, Dudra-Jastrzębska M, Pluta R, Czuczwar SJ. Anti-Epileptogenic Effects of Antiepileptic Drugs. Int J Mol Sci 2020; 21:ijms21072340. [PMID: 32231010 PMCID: PMC7178140 DOI: 10.3390/ijms21072340] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
Generally, the prevalence of epilepsy does not exceed 0.9% of the population and approximately 70% of epilepsy patients may be adequately controlled with antiepileptic drugs (AEDs). Moreover, status epilepticus (SE) or even a single seizure may produce neurodegeneration within the brain and SE has been recognized as one of acute brain insults leading to acquired epilepsy via the process of epileptogenesis. Two questions thus arise: (1) Are AEDs able to inhibit SE-induced neurodegeneration? and (2) if so, can a probable neuroprotective potential of particular AEDs stop epileptogenesis? An affirmative answer to the second question would practically point to the preventive potential of a given neuroprotective AED following acute brain insults. The available experimental data indicate that diazepam (at low and high doses), gabapentin, pregabalin, topiramate and valproate exhibited potent or moderate neuroprotective effects in diverse models of SE in rats. However, only diazepam (at high doses), gabapentin and pregabalin exerted some protective activity against acquired epilepsy (spontaneous seizures). As regards valproate, its effects on spontaneous seizures were equivocal. With isobolography, some supra-additive combinations of AEDs have been delineated against experimental seizures. One of such combinations, levetiracetam + topiramate proved highly synergistic in two models of seizures and this particular combination significantly inhibited epileptogenesis in rats following status SE. Importantly, no neuroprotection was evident. It may be strikingly concluded that there is no correlation between neuroprotection and antiepileptogenesis. Probably, preclinically verified combinations of AEDs may be considered for an anti-epileptogenic therapy.
Collapse
Affiliation(s)
- Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Agnieszka Konarzewska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Marzena Ułamek-Kozioł
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Monika Dudra-Jastrzębska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| |
Collapse
|
17
|
Klein P, Friedman A, Hameed MQ, Kaminski RM, Bar-Klein G, Klitgaard H, Koepp M, Jozwiak S, Prince DA, Rotenberg A, Twyman R, Vezzani A, Wong M, Löscher W. Repurposed molecules for antiepileptogenesis: Missing an opportunity to prevent epilepsy? Epilepsia 2020; 61:359-386. [PMID: 32196665 PMCID: PMC8317585 DOI: 10.1111/epi.16450] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Prevention of epilepsy is a great unmet need. Acute central nervous system (CNS) insults such as traumatic brain injury (TBI), cerebrovascular accidents (CVA), and CNS infections account for 15%-20% of all epilepsy. Following TBI and CVA, there is a latency of days to years before epilepsy develops. This allows treatment to prevent or modify postinjury epilepsy. No such treatment exists. In animal models of acquired epilepsy, a number of medications in clinical use for diverse indications have been shown to have antiepileptogenic or disease-modifying effects, including medications with excellent side effect profiles. These include atorvastatin, ceftriaxone, losartan, isoflurane, N-acetylcysteine, and the antiseizure medications levetiracetam, brivaracetam, topiramate, gabapentin, pregabalin, vigabatrin, and eslicarbazepine acetate. In addition, there are preclinical antiepileptogenic data for anakinra, rapamycin, fingolimod, and erythropoietin, although these medications have potential for more serious side effects. However, except for vigabatrin, there have been almost no translation studies to prevent or modify epilepsy using these potentially "repurposable" medications. We may be missing an opportunity to develop preventive treatment for epilepsy by not evaluating these medications clinically. One reason for the lack of translation studies is that the preclinical data for most of these medications are disparate in terms of types of injury, models within different injury type, dosing, injury-treatment initiation latencies, treatment duration, and epilepsy outcome evaluation mode and duration. This makes it difficult to compare the relative strength of antiepileptogenic evidence across the molecules, and difficult to determine which drug(s) would be the best to evaluate clinically. Furthermore, most preclinical antiepileptogenic studies lack information needed for translation, such as dose-blood level relationship, brain target engagement, and dose-response, and many use treatment parameters that cannot be applied clinically, for example, treatment initiation before or at the time of injury and dosing higher than tolerated human equivalent dosing. Here, we review animal and human antiepileptogenic evidence for these medications. We highlight the gaps in our knowledge for each molecule that need to be filled in order to consider clinical translation, and we suggest a platform of preclinical antiepileptogenesis evaluation of potentially repurposable molecules or their combinations going forward.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland
| | - Alon Friedman
- Departments of Physiology and Cell Biology, and Brain and Cognitive Science, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Departments of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Canada
| | - Mustafa Q. Hameed
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rafal M. Kaminski
- Neurosymptomatic Domains Section, Roche Pharma Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Guy Bar-Klein
- McKusick-Nathans Institute of Genetic Medicine, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henrik Klitgaard
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l’Alleud, Belgium
| | - Mathias Koepp
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Sergiusz Jozwiak
- Department of Pediatric Neurology, Warsaw Medical University, Warsaw, Poland
| | - David A. Prince
- Neurology and the Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Scientific Institute for Research and Health Care, Milan, Italy
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
18
|
Schidlitzki A, Bascuñana P, Srivastava PK, Welzel L, Twele F, Töllner K, Käufer C, Gericke B, Feleke R, Meier M, Polyak A, Ross TL, Gerhauser I, Bankstahl JP, Johnson MR, Bankstahl M, Löscher W. Proof-of-concept that network pharmacology is effective to modify development of acquired temporal lobe epilepsy. Neurobiol Dis 2019; 134:104664. [PMID: 31678583 DOI: 10.1016/j.nbd.2019.104664] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/08/2019] [Accepted: 10/30/2019] [Indexed: 10/25/2022] Open
Abstract
Epilepsy is a complex network phenomenon that, as yet, cannot be prevented or cured. We recently proposed network-based approaches to prevent epileptogenesis. For proof of concept we combined two drugs (levetiracetam and topiramate) for which in silico analysis of drug-protein interaction networks indicated a synergistic effect on a large functional network of epilepsy-relevant proteins. Using the intrahippocampal kainate mouse model of temporal lobe epilepsy, the drug combination was administered during the latent period before onset of spontaneous recurrent seizures (SRS). When SRS were periodically recorded by video-EEG monitoring after termination of treatment, a significant decrease in incidence and frequency of SRS was determined, indicating antiepileptogenic efficacy. Such efficacy was not observed following single drug treatment. Furthermore, a combination of levetiracetam and phenobarbital, for which in silico analysis of drug-protein interaction networks did not indicate any significant drug-drug interaction, was not effective to modify development of epilepsy. Surprisingly, the promising antiepileptogenic effect of the levetiracetam/topiramate combination was obtained in the absence of any significant neuroprotective or anti-inflammatory effects as indicated by multimodal brain imaging and histopathology. High throughput RNA-sequencing (RNA-seq) of the ipsilateral hippocampus of mice treated with the levetiracetam/topiramate combination showed that several genes that have been linked previously to epileptogenesis, were significantly differentially expressed, providing interesting entry points for future mechanistic studies. Overall, we have discovered a novel combination treatment with promise for prevention of epilepsy.
Collapse
Affiliation(s)
- Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | | | - Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Rahel Feleke
- Division of Brain Sciences, Imperial College London, London, UK
| | - Martin Meier
- Central Animal Facility & Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | | | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany; Central Animal Facility & Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
19
|
Gericke B, Brandt C, Theilmann W, Welzel L, Schidlitzki A, Twele F, Kaczmarek E, Anjum M, Hillmann P, Löscher W. Selective inhibition of mTORC1/2 or PI3K/mTORC1/2 signaling does not prevent or modify epilepsy in the intrahippocampal kainate mouse model. Neuropharmacology 2019; 162:107817. [PMID: 31654704 DOI: 10.1016/j.neuropharm.2019.107817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/26/2019] [Accepted: 10/18/2019] [Indexed: 12/23/2022]
Abstract
Dysregulation of the PI3K/Akt/mTOR pathway has been implicated in several brain disorders, including epilepsy. Rapamycin and similar compounds inhibit mTOR. complex 1 and have been reported to decrease seizures, delay seizure development, or prevent epileptogenesis in different animal models of genetic or acquired epilepsies. However, data for acquired epilepsy are inconsistent, which, at least in part, may be due to the poor brain penetration and long brain persistence of rapamycin and the fact that it blocks only one of the two cellular mTOR complexes. Here we examined the antiepileptogenic or disease-modifying effects of two novel, brain-permeable and well tolerated 1,3,5-triazine derivatives, the ATP-competitive mTORC1/2 inhibitor PQR620 and the dual pan-PI3K/mTORC1/2 inhibitor PQR530 in the intrahippocampal kainate mouse model, in which spontaneous seizures develop after status epilepticus (SE). Following kainate injection, the two compounds were administered over 2 weeks at doses previously been shown to block mTORC1/2 or PI3K/mTORC1/2 in the mouse brain. When spontaneous seizures were recorded by continuous (24/7) video-EEG recording starting 6 weeks after termination of treatment, no effects on incidence or frequency of seizures were observed. Drug treatment suppressed the epilepsy-induced activation of the PI3K/Akt/mTOR pathway in the hippocampus, but granule cell dispersion in the dentate gyrus was not prevented. When epilepsy-associated behavioral alterations were determined 12-14 weeks after kainate, mice pretreated with PQR620 or PQR530 exhibited reduced anxiety-related behavior in the light-dark box, indicating a disease-modifying effect. Overall, the data indicate that mTORC1/C2 or PI3K/mTORC1/C2 inhibition may not be an antiepileptogenic strategy for SE-induced epilepsy.
Collapse
Affiliation(s)
- Birthe Gericke
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Wiebke Theilmann
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Lisa Welzel
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Edith Kaczmarek
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Muneeb Anjum
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | | | - Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
20
|
Kwon JY, Jeon MT, Jung UJ, Kim DW, Moon GJ, Kim SR. Perspective: Therapeutic Potential of Flavonoids as Alternative Medicines in Epilepsy. Adv Nutr 2019; 10:778-790. [PMID: 31111873 PMCID: PMC6743823 DOI: 10.1093/advances/nmz047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/05/2019] [Accepted: 04/08/2019] [Indexed: 12/18/2022] Open
Abstract
Epilepsy is a chronic neurological disorder that affects many people worldwide. Temporal lobe epilepsy is the most common and most studied type of epilepsy, but the pathological mechanisms underlying this condition are poorly understood. More than 20 antiepileptic drugs (AEDs) have been developed and used for the treatment of epilepsy; however, 30% of patients still experience uncontrolled epilepsy and associated comorbidities, which impair their quality of life. In addition, various side effects have been reported for AEDs, such as drowsiness, unsteadiness, dizziness, blurred or double vision, tremor (shakiness), greater risk of infections, bruising, and bleeding. Thus, critical medical needs remain unmet for patients with uncontrolled epilepsy. Flavonoids belong to a subclass of polyphenols that are widely present in fruits, vegetables, and certain beverages. Recently, many studies have reported that some flavonoids elicit various beneficial effects in patients with epilepsy without causing the side effects associated with conventional medical therapies. Moreover, flavonoids may have a property of regulating microRNA expression associated with inflammation and cell survival. These findings suggest that flavonoids, which are more effective but impose fewer adverse effects than conventional AEDs, could be used in the treatment of epilepsy.
Collapse
Affiliation(s)
- Jae Young Kwon
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Min-Tae Jeon
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science,Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Gyeong Joon Moon
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea,Address correspondence to GJM (e-mail: )
| | - Sang Ryong Kim
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea,Address correspondence to SRK (e-mail: )
| |
Collapse
|
21
|
Jiang X, Lupien-Meilleur A, Tazerart S, Lachance M, Samarova E, Araya R, Lacaille JC, Rossignol E. Remodeled cortical inhibition prevents motor seizures in generalized epilepsy. Ann Neurol 2019; 84:436-451. [PMID: 30048010 DOI: 10.1002/ana.25301] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/12/2018] [Accepted: 07/21/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Deletions of CACNA1A, encoding the α1 subunit of CaV 2.1 channels, cause epilepsy with ataxia in humans. Whereas the deletion of Cacna1a in γ-aminobutyric acidergic (GABAergic) interneurons (INs) derived from the medial ganglionic eminence (MGE) impairs cortical inhibition and causes generalized seizures in Nkx2.1Cre ;Cacna1ac/c mice, the targeted deletion of Cacna1a in somatostatin-expressing INs (SOM-INs), a subset of MGE-derived INs, does not result in seizures, indicating a crucial role of parvalbumin-expressing (PV) INs. Here we identify the cellular and network consequences of Cacna1a deletion specifically in PV-INs. METHODS We generated PVCre ;Cacna1ac/c mutant mice carrying a conditional Cacna1a deletion in PV neurons and evaluated the cortical cellular and network outcomes of this mutation by combining immunohistochemical assays, in vitro electrophysiology, 2-photon imaging, and in vivo video-electroencephalographic recordings. RESULTS PVCre ;Cacna1ac/c mice display reduced cortical perisomatic inhibition and frequent absences but only rare motor seizures. Compared to Nkx2.1Cre ;Cacna1ac/c mice, PVCre ;Cacna1ac/c mice have a net increase in cortical inhibition, with a gain of dendritic inhibition through sprouting of SOM-IN axons, largely preventing motor seizures. This beneficial compensatory remodeling of cortical GABAergic innervation is mTORC1-dependent and its inhibition with rapamycin leads to a striking increase in motor seizures. Furthermore, we show that a direct chemogenic activation of cortical SOM-INs prevents motor seizures in a model of kainate-induced seizures. INTERPRETATION Our findings provide novel evidence suggesting that the remodeling of cortical inhibition, with an mTOR-dependent gain of dendritic inhibition, determines the seizure phenotype in generalized epilepsy and that mTOR inhibition can be detrimental in epilepsies not primarily due to mTOR hyperactivation. Ann Neurol 2018;84:436-451.
Collapse
Affiliation(s)
- Xiao Jiang
- Sainte-Justine University Hospital Research Center.,Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| | | | - Sabrina Tazerart
- Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| | | | - Elena Samarova
- Sainte-Justine University Hospital Research Center.,Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| | - Roberto Araya
- Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| | - Jean-Claude Lacaille
- Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| | - Elsa Rossignol
- Sainte-Justine University Hospital Research Center.,Department of Neurosciences and the Central Nervous System Research Group, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Löscher W. The holy grail of epilepsy prevention: Preclinical approaches to antiepileptogenic treatments. Neuropharmacology 2019; 167:107605. [PMID: 30980836 DOI: 10.1016/j.neuropharm.2019.04.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
A variety of acute brain insults can induce epileptogenesis, a complex process that results in acquired epilepsy. Despite advances in understanding mechanisms of epileptogenesis, there is currently no approved treatment that prevents the development or progression of epilepsy in patients at risk. The current concept of epileptogenesis assumes a window of opportunity following acute brain insults that allows intervention with preventive treatment. Recent results suggest that injury-induced epileptogenesis can be a much more rapid process than previously thought, suggesting that the 'therapeutic window' may only be open for a brief period, as in stroke therapy. However, experimental data also suggest a second, possibly delayed process ("secondary epileptogenesis") that influences the progression and refractoriness of the epileptic state over time, allowing interfering with this process even after onset of epilepsy. In this review, both methodological issues in preclinical drug development and novel targets for antiepileptogenesis will be discussed. Several promising drugs that either prevent epilepsy (antiepileptogenesis) or slow epilepsy progression and alleviate cognitive or behavioral comorbidities of epilepsy (disease modification) have been described in recent years, using diverse animal models of acquired epilepsy. Promising agents include TrkB inhibitors, losartan, statins, isoflurane, anti-inflammatory and anti-oxidative drugs, the SV2A modulator levetiracetam, and epigenetic interventions. Research on translational target validity and on prognostic biomarkers that can be used to stratify patients (or experimental animals) at high risk of developing epilepsy will hopefully soon lead to proof-of-concept clinical trials with the most promising drugs, which will be essential to make prevention of epilepsy a reality. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
23
|
Jeong KH, Kim SH, Choi YH, Cho I, Kim WJ. Increased expression of WNK3 in dispersed granule cells in hippocampal sclerosis of mesial temporal lobe epilepsy patients. Epilepsy Res 2018; 147:58-61. [PMID: 30253317 DOI: 10.1016/j.eplepsyres.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/22/2018] [Accepted: 09/14/2018] [Indexed: 11/17/2022]
Abstract
Granule cell dispersion (GCD) is a common neuropathological feature of hippocampal sclerosis (HS) in patients with temporal lobe epilepsy (TLE). However, the underlying molecular mechanism of GCD formation remains unclear. The present study aimed to investigate the expressional changes of With No Lysine protein kinase subtype 3 (WNK3), a molecule upstream of cation-chloride cotransporters with reciprocal expression in sclerosed hippocampus of TLE patients. Using immunofluorescence staining, we analyzed WNK3 immunoreactivity in hippocampal specimens from histologically normal controls and TLE patients with HS. Our results showed that WNK3 expression was significantly increased in dispersed granule neurons in hippocampal tissues from patients with TLE compared with histologically normal hippocampus. These findings demonstrate a potential association between an increased expression of WNK3 and GCD formation during the chronic phase of epilepsy. Controlling WNK3 expression may thus be a novel therapeutic target in epileptogenesis.
Collapse
Affiliation(s)
- Kyoung Hoon Jeong
- Department of Neurology and Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Yun Ho Choi
- Department of Neurology, Incheon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Incheon, Republic of Korea
| | - Inja Cho
- Department of Neurology and Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Won-Joo Kim
- Department of Neurology and Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Danzer SC. Contributions of Adult-Generated Granule Cells to Hippocampal Pathology in Temporal Lobe Epilepsy: A Neuronal Bestiary. Brain Plast 2018; 3:169-181. [PMID: 30151341 PMCID: PMC6091048 DOI: 10.3233/bpl-170056] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hippocampal neurogenesis continues throughout life in mammals – including humans. During the development of temporal lobe epilepsy, newly-generated hippocampal granule cells integrate abnormally into the brain. Abnormalities include ectopic localization of newborn cells, de novo formation of abnormal basal dendrites, and disruptions of the apical dendritic tree. Changes in granule cell position and dendritic structure fundamentally alter the types of inputs these cells are able to receive, as well as the relative proportions of remaining inputs. Dendritic abnormalities also create new pathways for recurrent excitation in the hippocampus. These abnormalities are hypothesized to contribute to the development of epilepsy, and may underlie cognitive disorders associated with the disease as well. To test this hypothesis, investigators have used pharmacological and genetic strategies in animal models to alter neurogenesis rates, or ablate the newborn cells outright. While findings are mixed and many unanswered questions remain, numerous studies now demonstrate that ablating newborn granule cells can have disease modifying effects in epilepsy. Taken together, findings provide a strong rationale for continued work to elucidate the role of newborn granule cells in epilepsy: both to understand basic mechanisms underlying the disease, and as a potential novel therapy for epilepsy.
Collapse
Affiliation(s)
- Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
25
|
Brandt C, Hillmann P, Noack A, Römermann K, Öhler LA, Rageot D, Beaufils F, Melone A, Sele AM, Wymann MP, Fabbro D, Löscher W. The novel, catalytic mTORC1/2 inhibitor PQR620 and the PI3K/mTORC1/2 inhibitor PQR530 effectively cross the blood-brain barrier and increase seizure threshold in a mouse model of chronic epilepsy. Neuropharmacology 2018; 140:107-120. [PMID: 30081001 DOI: 10.1016/j.neuropharm.2018.08.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
The mTOR signaling pathway has emerged as a possible therapeutic target for epilepsy. Clinical trials have shown that mTOR inhibitors such as everolimus reduce seizures in tuberous sclerosis complex patients with intractable epilepsy. Furthermore, accumulating preclinical data suggest that mTOR inhibitors may have anti-seizure or anti-epileptogenic actions in other types of epilepsy. However, the chronic use of rapalogs such as everolimus is limited by poor tolerability, particularly by immunosuppression, poor brain penetration and induction of feedback loops which might contribute to their limited therapeutic efficacy. Here we describe two novel, brain-permeable and well tolerated small molecule 1,3,5-triazine derivatives, the catalytic mTORC1/C2 inhibitor PQR620 and the dual pan-PI3K/mTOR inhibitor PQR530. These derivatives were compared with the mTORC1 inhibitors rapamycin and everolimus as well as the anti-seizure drugs phenobarbital and levetiracetam. The anti-seizure potential of these compounds was determined by evaluating the electroconvulsive seizure threshold in normal and epileptic mice. Rapamycin and everolimus only poorly penetrated into the brain (brain:plasma ratio 0.0057 for rapamycin and 0.016 for everolimus). In contrast, the novel compounds rapidly entered the brain, reaching brain:plasma ratios of ∼1.6. Furthermore, they significantly decreased phosphorylation of S6 ribosomal protein in the hippocampus of normal and epileptic mice, demonstrating effective mTOR inhibition. PQR620 and PQR530 significantly increased seizure threshold at tolerable doses. The effect of PQR620 was more marked in epileptic vs. nonepileptic mice, matching the efficacy of levetiracetam. Overall, the novel compounds described here have the potential to overcome the disadvantages of rapalogs for treatment of epilepsy and mTORopathies directly connected to mutations in the mTOR signaling cascade.
Collapse
Affiliation(s)
- Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | | | - Andreas Noack
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Leon A Öhler
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Denise Rageot
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Anna Melone
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alexander M Sele
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
26
|
Lee JM, Hong J, Moon GJ, Jung UJ, Won SY, Kim SR. Morin Prevents Granule Cell Dispersion and Neurotoxicity via Suppression of mTORC1 in a Kainic Acid-induced Seizure Model. Exp Neurobiol 2018; 27:226-237. [PMID: 30022874 PMCID: PMC6050420 DOI: 10.5607/en.2018.27.3.226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/21/2022] Open
Abstract
An abnormal reorganization of the dentate gyrus and neurotoxic events are important phenotypes in the hippocampus of patients with temporal lobe epilepsy (TLE). The effects of morin, a bioflavonoid constituent of many herbs and fruits, on epileptic seizures have not yet been elucidated, though its beneficial effects, such as its anti-inflammatory and neuroprotective properties, are well-described in various neurodegenerative diseases. In the present study, we investigated whether treatment with morin hydrate (MH) can reduce the susceptibility to seizures, granule cell dispersion (GCD), mammalian target of rapamycin complex 1 (mTORC1) activity, and the increases in the levels of apoptotic molecules and inflammatory cytokines in the kainic acid (KA)-induced seizure mouse model. Our results showed that oral administration of MH could reduce susceptibility to seizures and lead to the inhibition of GCD and mTORC1 activity in the KA-treated hippocampus. Moreover, treatment with MH significantly reduced the increased levels of apoptotic signaling molecules and pro-inflammatory mediators in the KA-treated hippocampus compared with control mice, suggesting a neuroprotective role. Therefore, these results suggest that morin has a therapeutic potential against epilepsy through its abilities to inhibit GCD and neurotoxic events in the in vivo hippocampus.
Collapse
Affiliation(s)
- Ji Min Lee
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Jungwan Hong
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea
| | - Gyeong Joon Moon
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Korea
| | - So-Yoon Won
- Department of Biochemistry and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
27
|
Godale CM, Danzer SC. Signaling Pathways and Cellular Mechanisms Regulating Mossy Fiber Sprouting in the Development of Epilepsy. Front Neurol 2018; 9:298. [PMID: 29774009 PMCID: PMC5943493 DOI: 10.3389/fneur.2018.00298] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/17/2018] [Indexed: 02/04/2023] Open
Abstract
The sprouting of hippocampal dentate granule cell axons, termed mossy fibers, into the dentate inner molecular layer is one of the most consistent findings in tissue from patients with mesial temporal lobe epilepsy. Decades of research in animal models have revealed that mossy fiber sprouting creates de novo recurrent excitatory connections in the hippocampus, fueling speculation that the pathology may drive temporal lobe epileptogenesis. Conducting definitive experiments to test this hypothesis, however, has been challenging due to the difficulty of dissociating this sprouting from the many other changes occurring during epileptogenesis. The field has been largely driven, therefore, by correlative data. Recently, the development of powerful transgenic mouse technologies and the discovery of novel drug targets has provided new tools to assess the role of mossy fiber sprouting in epilepsy. We can now selectively manipulate hippocampal granule cells in rodent epilepsy models, providing new insights into the granule cell subpopulations that participate in mossy fiber sprouting. The cellular pathways regulating this sprouting are also coming to light, providing new targets for pharmacological intervention. Surprisingly, many investigators have found that blocking mossy fiber sprouting has no effect on seizure occurrence, while seizure frequency can be reduced by treatments that have no effect on this sprouting. These results raise new questions about the role of mossy fiber sprouting in epilepsy. Here, we will review these findings with particular regard to the contributions of new granule cells to mossy fiber sprouting and the regulation of this sprouting by the mTOR signaling pathway.
Collapse
Affiliation(s)
- Christin M Godale
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States.,Department of Anesthesia, University of Cincinnati, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
28
|
Nguyen DL, Wimberley C, Truillet C, Jego B, Caillé F, Pottier G, Boisgard R, Buvat I, Bouilleret V. Longitudinal positron emission tomography imaging of glial cell activation in a mouse model of mesial temporal lobe epilepsy: Toward identification of optimal treatment windows. Epilepsia 2018; 59:1234-1244. [PMID: 29672844 DOI: 10.1111/epi.14083] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Mesiotemporal lobe epilepsy is the most common type of drug-resistant partial epilepsy, with a specific history that often begins with status epilepticus due to various neurological insults followed by a silent period. During this period, before the first seizure occurs, a specific lesion develops, described as unilateral hippocampal sclerosis (HS). It is still challenging to determine which drugs, administered at which time point, will be most effective during the formation of this epileptic process. Neuroinflammation plays an important role in pathophysiological mechanisms in epilepsy, and therefore brain inflammation biomarkers such as translocator protein 18 kDa (TSPO) can be potent epilepsy biomarkers. TSPO is associated with reactive astrocytes and microglia. A unilateral intrahippocampal kainate injection mouse model can reproduce the defining features of human temporal lobe epilepsy with unilateral HS and the pattern of chronic pharmacoresistant temporal seizures. We hypothesized that longitudinal imaging using TSPO positron emission tomography (PET) with 18 F-DPA-714 could identify optimal treatment windows in a mouse model during the formation of HS. METHODS The model was induced into the right dorsal hippocampus of male C57/Bl6 mice. Micro-PET/computed tomographic scanning was performed before model induction and along the development of the HS at 7 days, 14 days, 1 month, and 6 months. In vitro autoradiography and immunohistofluorescence were performed on additional mice at each time point. RESULTS TSPO PET uptake reached peak at 7 days and mostly related to microglial activation, whereas after 14 days, reactive astrocytes were shown to be the main cells expressing TSPO, reflected by a continuing increased PET uptake. SIGNIFICANCE TSPO-targeted PET is a highly potent longitudinal biomarker of epilepsy and could be of interest to determine the therapeutic windows in epilepsy and to monitor response to treatment.
Collapse
Affiliation(s)
- Duc-Loc Nguyen
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Catriona Wimberley
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Charles Truillet
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Benoit Jego
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Fabien Caillé
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Géraldine Pottier
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Raphaël Boisgard
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Irène Buvat
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France
| | - Viviane Bouilleret
- In Vivo Molecular Imaging Laboratory (IMIV), French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), French National Center for Scientific Research (CNRS), Paris Saclay University, Frédéric Joliot Hospital service, Orsay, France.,Neurophysiology and Epilepsy Unit, Bicêtre Hospital, Public Hospitals of Paris (AP-HP), France
| |
Collapse
|
29
|
Calderon-Garcidueñas AL, Mathon B, Lévy P, Bertrand A, Mokhtari K, Samson V, Thuriès V, Lambrecq V, Nguyen VHM, Dupont S, Adam C, Baulac M, Clémenceau S, Duyckaerts C, Navarro V, Bielle F. New clinicopathological associations and histoprognostic markers in ILAE types of hippocampal sclerosis. Brain Pathol 2018; 28:644-655. [PMID: 29476662 DOI: 10.1111/bpa.12596] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 01/06/2018] [Accepted: 01/27/2018] [Indexed: 12/30/2022] Open
Abstract
Mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE-HS) is a heterogeneous syndrome. Surgery results in seizure freedom for most pharmacoresistant patients, but the epileptic and cognitive prognosis remains variable. The 2013 International League Against Epilepsy (ILAE) histopathological classification of hippocampal sclerosis (HS) has fostered research to understand MTLE-HS heterogeneity. We investigated the associations between histopathological features (ILAE types, hypertrophic CA4 neurons, granule cell layer alterations, CD34 immunopositive cells) and clinical features (presurgical history, postsurgical outcome) in a monocentric series of 247 MTLE-HS patients treated by surgery. NeuN, GFAP and CD34 immunostainings and a double independent pathological examination were performed. 186 samples were type 1, 47 type 2, 7 type 3 and 7 samples were gliosis only but no neuronal loss (noHS). In the type 1, hypertrophic CA4 neurons were associated with a worse postsurgical outcome and granule cell layer duplication was associated with generalized seizures and episodes of status epilepticus. In the type 2, granule cell layer duplication was associated with generalized seizures. CD34+ stellate cells were more frequent in the type 2, type 3 and in noHS. These cells had a Nestin and SOX2 positive, immature neural immunophenotype. Patients with nodules of CD34+ cells had more frequent dysmnesic auras. CD34+ stellate cells in scarce pattern were associated with higher ratio of normal MRI and of stereo-electroencephalographic studies. CD34+ cells were associated with a trend for a better postsurgical outcome. Among CD34+ cases, we proposed a new entity of BRAF V600E positive HS and we described three hippocampal multinodular and vacuolating neuronal tumors. To conclude, our data identified new clinicopathological associations with ILAE types. They showed the prognostic value of CA4 hypertrophic neurons. They highlighted CD34+ stellate cells and BRAF V600E as biomarkers to further decipher MTLE-HS heterogeneity.
Collapse
Affiliation(s)
- Ana Laura Calderon-Garcidueñas
- Department of Neuropathology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Institute of Forensic Medicine, Universidad Veracruzana, Boca del Río, Mexico
| | - Bertrand Mathon
- Department of Neurosurgery, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Sorbonne University, UPMC, Univ Paris 06, Paris, France
| | - Pierre Lévy
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,UPMC and Inserm UMR S 1136 (EPAR team), Département de Santé Publique, Hôpital Tenon, Groupe Hospitalier Universitaire de l'Est Parisien, AP-HP, Paris, France
| | - Anne Bertrand
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Inria Paris, Aramis project-team, Paris, France.,Department of Radiology, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Karima Mokhtari
- Department of Neuropathology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France
| | - Véronique Samson
- Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Valérie Thuriès
- Department of Neuropathology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Virginie Lambrecq
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Vi-Huong Michel Nguyen
- Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Sophie Dupont
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Department of Rehabilitation, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Claude Adam
- Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Michel Baulac
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Stéphane Clémenceau
- Department of Neurosurgery, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Charles Duyckaerts
- Department of Neuropathology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France
| | - Vincent Navarro
- Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France.,Department of Epileptology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France
| | - Franck Bielle
- Department of Neuropathology, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Paris, France.,Sorbonne University, UPMC, Univ Paris 06, Paris, France.,Brain and Spine Institute (ICM; INSERM, UMRS 1127; CNRS, UMR 7225), Paris, France
| |
Collapse
|
30
|
Tai XY, Bernhardt B, Thom M, Thompson P, Baxendale S, Koepp M, Bernasconi N. Review: Neurodegenerative processes in temporal lobe epilepsy with hippocampal sclerosis: Clinical, pathological and neuroimaging evidence. Neuropathol Appl Neurobiol 2018; 44:70-90. [DOI: 10.1111/nan.12458] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022]
Affiliation(s)
- X. Y. Tai
- Division of Neuropathology and Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - B. Bernhardt
- Neuroimaging of Epilepsy Laboratory; McConnell Brain Imaging Centre; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Multimodal Imaging and Connectome Analysis Lab; Montreal Neurological Institute; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - M. Thom
- Division of Neuropathology and Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - P. Thompson
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - S. Baxendale
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - M. Koepp
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - N. Bernasconi
- Neuroimaging of Epilepsy Laboratory; McConnell Brain Imaging Centre; Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| |
Collapse
|
31
|
Talos DM, Jacobs LM, Gourmaud S, Coto CA, Sun H, Lim KC, Lucas TH, Davis KA, Martinez-Lage M, Jensen FE. Mechanistic target of rapamycin complex 1 and 2 in human temporal lobe epilepsy. Ann Neurol 2018; 83:311-327. [PMID: 29331082 DOI: 10.1002/ana.25149] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Temporal lobe epilepsy (TLE) is a chronic epilepsy syndrome defined by seizures and progressive neurological disabilities, including cognitive impairments, anxiety, and depression. Here, human TLE specimens were investigated focusing on the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) and complex 2 (mTORC2) activities in the brain, given that both pathways may represent unique targets for treatment. METHODS Surgically resected hippocampal and temporal lobe samples from therapy-resistant TLE patients were analyzed by western blotting to quantify the expression of established mTORC1 and mTORC2 activity markers and upstream or downstream signaling pathways involving the two complexes. Histological and immunohistochemical techniques were used to assess hippocampal and neocortical structural abnormalities and cell-specific expression of individual biomarkers. Samples from patients with focal cortical dysplasia (FCD) type II served as positive controls. RESULTS We found significantly increased expression of phospho-mTOR (Ser2448), phospho-S6 (Ser235/236), phospho-S6 (Ser240/244), and phospho-Akt (Ser473) in TLE samples compared to controls, consistent with activation of both mTORC1 and mTORC2. Our work identified the phosphoinositide 3-kinase and Ras/extracellular signal-regulated kinase signaling pathways as potential mTORC1 and mTORC2 upstream activators. In addition, we found that overactive mTORC2 signaling was accompanied by induction of two protein kinase B-dependent prosurvival pathways, as evidenced by increased inhibitory phosphorylation of forkhead box class O3a (Ser253) and glycogen synthase kinase 3 beta (Ser9). INTERPRETATION Our data demonstrate that mTOR signaling is significantly dysregulated in human TLE, offering new targets for pharmacological interventions. Specifically, clinically available drugs that suppress mTORC1 without compromising mTOR2 signaling, such as rapamycin and its analogs, may represent a new group of antiepileptogenic agents in TLE patients. Ann Neurol 2018;83:311-327.
Collapse
Affiliation(s)
- Delia M Talos
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Leah M Jacobs
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Sarah Gourmaud
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Carlos A Coto
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Hongyu Sun
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA.,Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Kuei-Cheng Lim
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Timothy H Lucas
- Department of Neurosurgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Kathryn A Davis
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Maria Martinez-Lage
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Frances E Jensen
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
32
|
Naringenin ameliorates kainic acid-induced morphological alterations in the dentate gyrus in a mouse model of temporal lobe epilepsy. Neuroreport 2018; 27:1182-9. [PMID: 27584687 DOI: 10.1097/wnr.0000000000000678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Granule cell dispersion (GCD) in the dentate gyrus (DG) of the hippocampus is a morphological alteration characteristic of temporal lobe epilepsy. Recently, we reported that treatment with naringin, a flavonoid found in grapefruit and citrus fruits, reduced spontaneous recurrent seizures by inhibiting kainic acid (KA)-induced GCD and neuronal cell death in mouse hippocampus, suggesting that naringin might have beneficial effects for preventing epileptic events in the adult brain. However, it is still unclear whether the beneficial effects of naringin treatment are mediated by the metabolism of naringin into naringenin in the KA-treated hippocampus. To investigate this possibility, we evaluated whether intraperitoneal injections of naringenin could mimic naringin-induced effects against GCD caused by intrahippocampal KA injections in mice. Our results showed that treatment with naringenin delayed the onset of KA-induced seizures and attenuated KA-induced GCD by inhibiting activation of the mammalian target of rapamycin complex 1 in both neurons and reactive astrocytes in the DG. In addition, its administration attenuated the production of proinflammatory cytokines such as tumor necrosis tumor necrosis factor-α (TNFα) and interleukin-1β (IL-1β) from microglial activation in the DG following KA treatment. These results suggest that naringenin may be an active metabolite of naringin and help prevent the progression of epileptic insults in the hippocampus in vivo; therefore, naringenin may be a beneficial metabolite of naringin for the treatment of epilepsy.
Collapse
|
33
|
Neuberger EJ, Gupta A, Subramanian D, Korgaonkar AA, Santhakumar V. Converging early responses to brain injury pave the road to epileptogenesis. J Neurosci Res 2017; 97:1335-1344. [PMID: 29193309 DOI: 10.1002/jnr.24202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/19/2022]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal electrical activity in the brain, is one of the most prevalent brain disorders. Over two million people in the United States have been diagnosed with epilepsy and 3% of the general population will be diagnosed with it at some point in their lives. While most developmental epilepsies occur due to genetic predisposition, a class of "acquired" epilepsies results from a variety of brain insults. A leading etiological factor for epilepsy that is currently on the rise is traumatic brain injury (TBI), which accounts for up to 20% of all symptomatic epilepsies. Remarkably, the presence of an identified early insult that constitutes a risk for development of epilepsy provides a therapeutic window in which the pathological processes associated with brain injury can be manipulated to limit the subsequent development of recurrent seizure activity and epilepsy. Recent studies have revealed diverse pathologies, including enhanced excitability, activated immune signaling, cell death, and enhanced neurogenesis within a week after injury, suggesting a period of heightened adaptive and maladaptive plasticity. An integrated understanding of these processes and their cellular and molecular underpinnings could lead to novel targets to arrest epileptogenesis after trauma. This review attempts to highlight and relate the diverse early changes after trauma and their role in development of epilepsy and suggests potential strategies to limit neurological complications in the injured brain.
Collapse
Affiliation(s)
- Eric J Neuberger
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Akshay Gupta
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Deepak Subramanian
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Akshata A Korgaonkar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| |
Collapse
|
34
|
Kim S, Jung UJ, Oh YS, Jeon MT, Kim HJ, Shin WH, Hong J, Kim SR. Beneficial Effects of Silibinin Against Kainic Acid-induced Neurotoxicity in the Hippocampus in vivo. Exp Neurobiol 2017; 26:266-277. [PMID: 29093635 PMCID: PMC5661059 DOI: 10.5607/en.2017.26.5.266] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 10/05/2017] [Indexed: 02/04/2023] Open
Abstract
Silibinin, an active constituent of silymarin extracted from milk thistle, has been previously reported to confer protection to the adult brain against neurodegeneration. However, its effects against epileptic seizures have not been examined yet. In order to investigate the effects of silibinin against epileptic seizures, we used a relevant mouse model in which seizures are manifested as status epilepticus, induced by kainic acid (KA) treatment. Silibinin was injected intraperitoneally, starting 1 day before an intrahippocampal KA injection and continued daily until analysis of each experiment. Our results indicated that silibinin-treatment could reduce seizure susceptibility and frequency of spontaneous recurrent seizures (SRS) induced by KA administration, and attenuate granule cell dispersion (GCD), a morphological alteration characteristic of the dentate gyrus (DG) in temporal lobe epilepsy (TLE). Moreover, its treatment significantly reduced the aberrant levels of apoptotic, autophagic and pro-inflammatory molecules induced by KA administration, resulting in neuroprotection in the hippocampus. Thus, these results suggest that silibinin may be a beneficial natural compound for preventing epileptic events.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Korea
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| | - Min-Tae Jeon
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Hyung-Jun Kim
- Department of Neural Development and Disease, Department of Structure & Function of Neural Network, Korea Brain Research Institute, Daegu 41062, Korea
| | - Won-Ho Shin
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Jungwan Hong
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
35
|
Schidlitzki A, Twele F, Klee R, Waltl I, Römermann K, Bröer S, Meller S, Gerhauser I, Rankovic V, Li D, Brandt C, Bankstahl M, Töllner K, Löscher W. A combination of NMDA and AMPA receptor antagonists retards granule cell dispersion and epileptogenesis in a model of acquired epilepsy. Sci Rep 2017; 7:12191. [PMID: 28939854 PMCID: PMC5610327 DOI: 10.1038/s41598-017-12368-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/08/2017] [Indexed: 01/01/2023] Open
Abstract
Epilepsy may arise following acute brain insults, but no treatments exist that prevent epilepsy in patients at risk. Here we examined whether a combination of two glutamate receptor antagonists, NBQX and ifenprodil, acting at different receptor subtypes, exerts antiepileptogenic effects in the intrahippocampal kainate mouse model of epilepsy. These drugs were administered over 5 days following kainate. Spontaneous seizures were recorded by video/EEG at different intervals up to 3 months. Initial trials showed that drug treatment during the latent period led to higher mortality than treatment after onset of epilepsy, and further, that combined therapy with both drugs caused higher mortality at doses that appear safe when used singly. We therefore refined the combined-drug protocol, using lower doses. Two weeks after kainate, significantly less mice of the NBQX/ifenprodil group exhibited electroclinical seizures compared to vehicle controls, but this effect was lost at subsequent weeks. The disease modifying effect of the treatment was associated with a transient prevention of granule cell dispersion and less neuronal degeneration in the dentate hilus. These data substantiate the involvement of altered glutamatergic transmission in the early phase of epileptogenesis. Longer treatment with NBQX and ifenprodil may shed further light on the apparent temporal relationship between dentate gyrus reorganization and development of spontaneous seizures.
Collapse
Affiliation(s)
- Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Inken Waltl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sebastian Meller
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Vladan Rankovic
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Institute for Auditory Neuroscience at University Medical Center Göttingen & German Primate Center, Göttingen, Germany
| | - Dandan Li
- Center for Systems Neuroscience, 30559, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany.
- Center for Systems Neuroscience, 30559, Hannover, Germany.
| |
Collapse
|
36
|
Łukawski K, Andres-Mach M, Czuczwar M, Łuszczki JJ, Kruszyński K, Czuczwar SJ. Mechanisms of epileptogenesis and preclinical approach to antiepileptogenic therapies. Pharmacol Rep 2017; 70:284-293. [PMID: 29477036 DOI: 10.1016/j.pharep.2017.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/17/2017] [Accepted: 07/12/2017] [Indexed: 12/30/2022]
Abstract
The prevalence of epilepsy is estimated 5-10 per 1000 population and around 70% of patients with epilepsy can be sufficiently controlled by antiepileptic drugs (AEDs). Epileptogenesis is the process responsible for converting normal into an epileptic brain and mechanisms responsible include among others: inflammation, neurodegeneration, neurogenesis, neural reorganization and plasticity. Some AEDs may be antiepileptiogenic (diazepam, eslicarbazepine) but the correlation between neuroprotection and inhibition of epileptogenesis is not evident. Antiepileptogenic activity has been postulated for mTOR ligands, resveratrol and losartan. So far, clinical evidence gives some hope for levetiracetam as an AED inhibiting epileptogenesis in neurosurgical patients. Biomarkers for epileptogenesis are needed for the proper selection of patients for evaluation of potential antiepileptogenic compounds.
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland; Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Marta Andres-Mach
- Isobolographic Analysis Laboratory, Institute of Rural Health, Lublin, Poland
| | - Mirosław Czuczwar
- 2nd Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Isobolographic Analysis Laboratory, Institute of Rural Health, Lublin, Poland
| | | | - Stanisław J Czuczwar
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland; Department of Pathophysiology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
37
|
Switon K, Kotulska K, Janusz-Kaminska A, Zmorzynska J, Jaworski J. Molecular neurobiology of mTOR. Neuroscience 2017; 341:112-153. [PMID: 27889578 DOI: 10.1016/j.neuroscience.2016.11.017] [Citation(s) in RCA: 297] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/09/2016] [Accepted: 11/13/2016] [Indexed: 01/17/2023]
Abstract
Mammalian/mechanistic target of rapamycin (mTOR) is a serine-threonine kinase that controls several important aspects of mammalian cell function. mTOR activity is modulated by various intra- and extracellular factors; in turn, mTOR changes rates of translation, transcription, protein degradation, cell signaling, metabolism, and cytoskeleton dynamics. mTOR has been repeatedly shown to participate in neuronal development and the proper functioning of mature neurons. Changes in mTOR activity are often observed in nervous system diseases, including genetic diseases (e.g., tuberous sclerosis complex, Pten-related syndromes, neurofibromatosis, and Fragile X syndrome), epilepsy, brain tumors, and neurodegenerative disorders (Alzheimer's disease, Parkinson's disease, and Huntington's disease). Neuroscientists only recently began deciphering the molecular processes that are downstream of mTOR that participate in proper function of the nervous system. As a result, we are gaining knowledge about the ways in which aberrant changes in mTOR activity lead to various nervous system diseases. In this review, we provide a comprehensive view of mTOR in the nervous system, with a special focus on the neuronal functions of mTOR (e.g., control of translation, transcription, and autophagy) that likely underlie the contribution of mTOR to nervous system diseases.
Collapse
Affiliation(s)
- Katarzyna Switon
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, Warsaw 02-109, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Aleja Dzieci Polskich 20, Warsaw 04-730, Poland
| | | | - Justyna Zmorzynska
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, Warsaw 02-109, Poland
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, Warsaw 02-109, Poland.
| |
Collapse
|
38
|
Klingebiel M, Dinekov M, Köhler C. Analysis of ribosomal protein S6 baseline phosphorylation and effect of tau pathology in the murine brain and human hippocampus. Brain Res 2017; 1659:121-135. [PMID: 28119058 DOI: 10.1016/j.brainres.2017.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 01/17/2023]
Abstract
We examined the distribution pattern of the phosphorylated 40S ribosomal subunit protein S6, a downstream target of the mTOR pathway, in the brains of 24-months-old human tau transgenic pR5 mice, non-transgenic littermates and in human hippocampi. We studied baseline levels of phosphorylated S6 and a possible effect of tau pathology. S6 phosphorylated at Ser235/236 (pS6Ser235/236) or Ser240/244 (pS6Ser240/244) has been used as a read-out of mTOR activity in several studies. The mTOR pathway regulates a wide variety of cellular functions including cell growth, ribosome biosynthesis, translational control and autophagy. Its dysregulation might underlie the neurodegenerative pathology of Alzheimer's disease and other tauopathies. pS6Ser235/236 and pS6Ser240/244 immunoreactivity in the mouse brain were widespread and similar distributed, but intensive pS6Ser235/236 immunoreactivity was more selective, especially highlighting certain brainstem regions. In the human hippocampus mainly granulovacuolar inclusions in neurons displayed pS6Ser235/236 immunoreactivity. In contrast, a considerable number of neurons displayed pS6Ser240/244 immunoreactivity in the cytoplasm without labeling of granulovacuolar inclusions. Except for a tendency of lower numbers of intensely phosphorylated S6-positive neurons in pR5 mice, the pattern of distribution of pS6Ser235/236 and pS6Ser240/244 immunoreactivity was largely unchanged when compared with non-transgenic mice and also when human hippocampi from AD cases and controls were compared. Similar to pR5 mice most neurons with hyper-phosphorylated tau in human hippocampi displayed no or only weak labeling for phosphorylated S6, suggesting that phosphorylated S6 is not especially associated with pathological tau, but is rather a feature of unaffected neurons.
Collapse
Affiliation(s)
- Maria Klingebiel
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Maja Dinekov
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Christoph Köhler
- Institute II for Anatomy, Medical Faculty, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany.
| |
Collapse
|
39
|
Superimposing Status Epilepticus on Neuron Subset-Specific PTEN Haploinsufficient and Wild Type Mice Results in Long-term Changes in Behavior. Sci Rep 2016; 6:36559. [PMID: 27819284 PMCID: PMC5098193 DOI: 10.1038/srep36559] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/18/2016] [Indexed: 01/09/2023] Open
Abstract
We evaluated the effects of superimposing seizures on a genetic mutation with known involvement in both Autism Spectrum Disorder and in epilepsy. Neuron-subset specific (NS)-Pten heterozygous (HT) and wildtype (WT) adult mice received either intraperitoneal injections of kainic acid (20 mg/kg) to induce status epilepticus or the vehicle (saline). Animals then received a battery of behavioral tasks in order to evaluate activity levels, anxiety, repetitive-stereotyped behavior, social behavior, learning and memory. In the open field task, we found that HT mice after seizures showed a significant increase in total activity and total distance in the surround region of the open field. In the elevated plus maze task, we found that HT mice after seizures displayed increased total distance and velocity as compared to HT mice that did not undergo seizures and WT controls. In the social chamber test, we found the HT mice after seizures displayed an impairment in social behavior. These findings demonstrate that superimposing seizures on a genetic mutation can result in long-term alterations in activity and social behavior in mice.
Collapse
|
40
|
Kim SR. Control of Granule Cell Dispersion by Natural Materials Such as Eugenol and Naringin: A Potential Therapeutic Strategy Against Temporal Lobe Epilepsy. J Med Food 2016; 19:730-6. [PMID: 27404051 DOI: 10.1089/jmf.2016.3712] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The hippocampus is an important brain area where abnormal morphological characteristics are often observed in patients with temporal lobe epilepsy (TLE), typically showing the loss of the principal neurons in the CA1 and CA3 areas of the hippocampus. TLE is frequently associated with widening of the granule cell layer of the dentate gyrus (DG), termed granule cell dispersion (GCD), in the hippocampus, suggesting that the control of GCD with protection of hippocampal neurons may be useful for preventing and inhibiting epileptic seizures. We previously reported that eugenol (EUG), which is an essential component of medicinal herbs and has anticonvulsant activity, is beneficial for treating epilepsy through its ability to inhibit GCD via suppression of mammalian target of rapamycin complex 1 (mTORC1) activation in the hippocampal DG in a kainic acid (KA)-treated mouse model of epilepsy in vivo. In addition, we reported that naringin, a bioflavonoid in citrus fruits, could exert beneficial effects, such as antiautophagic stress and antineuroinflammation, in the KA mouse model of epilepsy, even though it was unclear whether naringin might also attenuate the seizure-induced morphological changes of GCD in the DG. Similar to the effects of EUG, we recently observed that naringin treatment significantly reduced KA-induced GCD and mTORC1 activation, which are both involved in epileptic seizures, in the hippocampus of mouse brain. Therefore, these observations suggest that the utilization of natural materials, which have beneficial properties such as inhibition of GCD formation and protection of hippocampal neurons, may be useful in developing a novel therapeutic agent against TLE.
Collapse
Affiliation(s)
- Sang Ryong Kim
- 1 School of Life Sciences, Kyungpook National University , Daegu, Korea.,2 BK21 plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Korea.,3 Brain Science and Engineering Institute, Kyungpook National University , Daegu, Korea
| |
Collapse
|
41
|
Naringin attenuates granule cell dispersion in the dentate gyrus in a mouse model of temporal lobe epilepsy. Epilepsy Res 2016; 123:6-10. [DOI: 10.1016/j.eplepsyres.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/18/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023]
|
42
|
Impact of rapamycin on status epilepticus induced hippocampal pathology and weight gain. Exp Neurol 2016; 280:1-12. [PMID: 26995324 DOI: 10.1016/j.expneurol.2016.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Growing evidence implicates the dentate gyrus in temporal lobe epilepsy (TLE). Dentate granule cells limit the amount of excitatory signaling through the hippocampus and exhibit striking neuroplastic changes that may impair this function during epileptogenesis. Furthermore, aberrant integration of newly-generated granule cells underlies the majority of dentate restructuring. Recently, attention has focused on the mammalian target of rapamycin (mTOR) signaling pathway as a potential mediator of epileptogenic change. Systemic administration of the mTOR inhibitor rapamycin has promising therapeutic potential, as it has been shown to reduce seizure frequency and seizure severity in rodent models. Here, we tested whether mTOR signaling facilitates abnormal development of granule cells during epileptogenesis. We also examined dentate inflammation and mossy cell death in the dentate hilus. To determine if mTOR activation is necessary for abnormal granule cell development, transgenic mice that harbored fluorescently-labeled adult-born granule cells were treated with rapamycin following pilocarpine-induced status epilepticus. Systemic rapamycin effectively blocked phosphorylation of S6 protein (a readout of mTOR activity) and reduced granule cell mossy fiber axon sprouting. However, the accumulation of ectopic granule cells and granule cells with aberrant basal dendrites was not significantly reduced. Mossy cell death and reactive astrocytosis were also unaffected. These data suggest that anti-epileptogenic effects of mTOR inhibition may be mediated by mechanisms other than inhibition of these common dentate pathologies. Consistent with this conclusion, rapamycin prevented pathological weight gain in epileptic mice, suggesting that rapamycin might act on central circuits or even peripheral tissues controlling weight gain in epilepsy.
Collapse
|
43
|
Drion CM, Borm LE, Kooijman L, Aronica E, Wadman WJ, Hartog AF, van Vliet EA, Gorter JA. Effects of rapamycin and curcumin treatment on the development of epilepsy after electrically induced status epilepticus in rats. Epilepsia 2016; 57:688-97. [DOI: 10.1111/epi.13345] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Cato M. Drion
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Lars E. Borm
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Lieneke Kooijman
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Eleonora Aronica
- Department (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Epilepsy Institute in the Netherlands; Heemstede The Netherlands
| | - Wytse J. Wadman
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Aloysius F. Hartog
- Van‘t Hoff Institute for Molecular Sciences; University of Amsterdam; Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Department (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jan A. Gorter
- Center for Neuroscience; Swammerdam Institute for Life Sciences; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
44
|
Kourdougli N, Varpula S, Chazal G, Rivera C. Detrimental effect of post Status Epilepticus treatment with ROCK inhibitor Y-27632 in a pilocarpine model of temporal lobe epilepsy. Front Cell Neurosci 2015; 9:413. [PMID: 26557054 PMCID: PMC4615811 DOI: 10.3389/fncel.2015.00413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/28/2015] [Indexed: 01/18/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults where 20-30% of the patients are refractory to currently available anti-epileptic drugs. The RhoA/Rho-kinase signaling pathway activation has been involved in inflammatory responses, neurite outgrowth and neuronal death under pathological conditions such as epileptic insults. Acute preventive administration of ROCK inhibitor has been reported to have beneficial outcomes in Status Epilepticus (SE) epilepsy. In the present study, we evaluate the effect of chronic post SE treatment with the ROCK inhibitor Y-27632 in a rat pilocarpine model of TLE. We used chronic i.p. injections of Y-27632 for 5 days in 6 week old control rats or rats subjected to pilocarpine treatment as a model of TLE. Surprisingly, our findings demonstrate that a systemic administration of Y-27632 in pilocarpine-treated rats increases neuronal death in the CA3 region and ectopic recurrent mossy fiber sprouting (rMFS) in the dentate gyrus of the hippocampal formation. Interestingly, we found that chronic treatment with Y-27632 exacerbates the down-regulation and pathological distribution of the K(+)-Cl(-) cotransporter KCC2, thus providing a putative mechanism for post SE induced neuronal death. The involvement of astrogliosis in this mechanism appears to be intricate as ROCK inhibition reduces reactive astrogliosis in pilocarpine rats. Conversely, in control rats, chronic Y-27632 treatment increases astrogliosis. Together, our findings suggest that Y-27632 has a detrimental effect when chronically used post SE in a rat pilocarpine model of TLE.
Collapse
Affiliation(s)
- Nazim Kourdougli
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
| | - Saara Varpula
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Genevieve Chazal
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
| | - Claudio Rivera
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| |
Collapse
|
45
|
Jeong KH, Lee DS, Kim SR. Effects of eugenol on granule cell dispersion in a mouse model of temporal lobe epilepsy. Epilepsy Res 2015. [DOI: 10.1016/j.eplepsyres.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|