51
|
Nguyen LH, Mahadeo T, Bordey A. mTOR Hyperactivity Levels Influence the Severity of Epilepsy and Associated Neuropathology in an Experimental Model of Tuberous Sclerosis Complex and Focal Cortical Dysplasia. J Neurosci 2019; 39:2762-2773. [PMID: 30700531 PMCID: PMC6445990 DOI: 10.1523/jneurosci.2260-18.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/28/2023] Open
Abstract
Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are focal malformations of cortical development (FMCDs) that are highly associated with intractable epilepsy. TSC and FCD are mTORopathies caused by a spectrum of pathogenic variants in the mechanistic target of rapamycin (mTOR) pathway genes leading to differential activation of mTOR signaling. However, whether the degree of mTOR hyperactivity influences disease severity remains unclear. Here, we examined the effects of differential mTOR hyperactivity levels on epilepsy and associated neuropathology in a mouse model of TSC and FCD. Constitutively active Rheb (RhebCA), the canonical activator of mTOR complex 1 (mTORC1), was expressed in mouse embryos of either sex via in utero electroporation at low, intermediate, and high concentrations to induce different mTORC1 activity levels in developing cortical neurons. We found that RhebCA expression induced mTORC1 hyperactivation and increased neuronal soma size and misplacement in a dose-dependent manner. No seizures were detected in the low RhebCA mice, whereas the intermediate and high RhebCA mice displayed spontaneous, recurrent seizures that significantly increased with higher RhebCA concentrations. Seizures were associated with a global increase in microglial activation that was notably higher in the regions containing RhebCA-expressing neurons. These data demonstrate that neuronal mTOR hyperactivity levels influence the severity of epilepsy and associated neuropathology in experimental TSC and FCD. Overall, these findings highlight the importance of evaluating the outcome of individual variants on mTOR activity levels and support personalized medicine strategies based on patient variants and mTOR activity level for TSC, FCD, and potentially other mTORopathies.SIGNIFICANCE STATEMENT Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are epileptogenic cortical malformations caused by pathogenic variants in mechanistic target of rapamycin (mTOR) pathway genes leading to differential mTOR hyperactivation. Here, we present novel findings that neuronal mTOR hyperactivity levels correlate with the severity of epilepsy and associated neuropathology in a mouse model of TSC and FCD. Our findings suggest the need to evaluate the outcome of individual variants on mTOR activity levels in clinical assessments and support personalized medicine strategies based on patient variants and mTOR activity level. Additionally, we present useful modifications to a previously described mouse model of TSC and FCD that allows for titration of seizure frequency and generation of a mild to severe epilepsy phenotype as applicable for preclinical drug testing and mechanistic studies.
Collapse
Affiliation(s)
| | | | - Angélique Bordey
- Department of Neurosurgery, and
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
52
|
Mazumder AG, Patial V, Singh D. Mycophenolate mofetil contributes to downregulation of the hippocampal interleukin type 2 and 1β mediated PI3K/AKT/mTOR pathway hyperactivation and attenuates neurobehavioral comorbidities in a rat model of temporal lobe epilepsy. Brain Behav Immun 2019; 75:84-93. [PMID: 30243822 DOI: 10.1016/j.bbi.2018.09.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 08/13/2018] [Accepted: 09/19/2018] [Indexed: 12/24/2022] Open
Abstract
The role of neuroinflammatory mediators has been well established in the pathogenesis of temporal lobe epilepsy (TLE) and associated neurobehavioral comorbidities. Mycophenolate mofetil (MMF) is commonly used as an immunosuppressant in organ transplantations. Its neuroprotective effect is well explored in different preclinical and clinical studies. The present study was designed to investigate the effect of MMF in rat model of lithium pilocarpine (LiPc)-induced spontaneous recurrent seizures and its associated neurobehavioral comorbidities. MMF treatment showed a dose-dependent decrease in seizure severity and reduced aggression in epileptic rats. There was marked improvement in spatial and recognition memory functions, along with substantial decrease in depression-like behavior in MMF treated epileptic rats. There was considerable decrease in mossy fiber sprouting in the dentate gyrus and the cornu ammonis 3 regions of the hippocampus, along with reduction in neuronal death in the treated groups. Furthermore, the hippocampal mRNA level of IL-1β, IL-2, PI3K, AKT, HIF-1α, RAPTOR, mTOR, Rps6kb1 and Rps6 was found to be decreased in MMF treated animals. mTOR, S6, pS6 and GFAP protein expression was decreased, whereas NeuN was increased in the rat hippocampus of the treated animals. The results concluded that MMF suppress recurrent seizures, and improves its associated behavioral impairments and cognitive deficit in rat model of TLE. The observed effects of MMF be correlated with the inhibition of IL-2 and IL-1β linked PI3K/AKT/mTOR signaling pathway hyperactivation.
Collapse
Affiliation(s)
- Arindam Ghosh Mazumder
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India.
| |
Collapse
|
53
|
Mizuguchi M, Ikeda H, Kagitani-Shimono K, Yoshinaga H, Suzuki Y, Aoki M, Endo M, Yonemura M, Kubota M. Everolimus for epilepsy and autism spectrum disorder in tuberous sclerosis complex: EXIST-3 substudy in Japan. Brain Dev 2019; 41:1-10. [PMID: 30060984 DOI: 10.1016/j.braindev.2018.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/28/2018] [Accepted: 07/09/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Epilepsy and autism spectrum disorder (ASD) are the common neurological manifestations of tuberous sclerosis complex (TSC). EXIST-3 study has recently demonstrated that everolimus reduces seizures in patients with TSC and refractory epilepsy. Here we report the efficacy and safety of everolimus for treatment-refractory seizures in Japanese patients of EXIST-3, along with the exploratory analysis evaluating the everolimus effect on comorbid ASD symptoms in these patients. METHODS Primary endpoint was change in seizure frequency from baseline defined as response rate (≥50% reduction) and median percentage reduction in the seizure frequency. Pervasive Developmental Disorders Autism Society Japan Rating Scale (PARS) scores were assessed at baseline and at week-18 for ASD symptoms. RESULTS Overall, 35 Japanese patients were randomized to everolimus low-exposure (LE; n = 10), everolimus high-exposure (HE; n = 14), or placebo (n = 11). The response rate was 30.0% and 28.6% versus 0% with the everolimus LE and HE versus placebo arm, respectively. Similarly, the median percentage reduction in seizure frequency was 6.88% and 38.06% versus -6.67%. Stomatitis was the most frequently reported adverse event (everolimus LE, 100%; HE, 78.6%; placebo, 9.1%). Four of 11 patients with ASD in the everolimus arms and 1 of 8 patients with ASD in the placebo arm showed ≥5 point decrease in PARS scores. CONCLUSIONS Adjunctive everolimus treatment improved seizure frequency with a tolerable safety relative to placebo among 35 Japanese patients with TSC-associated refractory seizures, consistent with the results of overall EXIST-3 study involving 366 patients. A favorable trend towards the improvement of ASD symptoms was observed.
Collapse
Affiliation(s)
- Masashi Mizuguchi
- Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Hiroko Ikeda
- Department of Pediatrics, Shizuoka Institute of Epilepsy and Neurological Disorders, NHO, Shizuoka, Japan
| | | | - Harumi Yoshinaga
- Department of Child Neurology, Okayama University Hospital, Okayama, Japan; Department of Severely Disabled Children Center, Minami Okayama Medical Center, Okayama, Japan
| | - Yasuhiro Suzuki
- Department of Pediatric Neurology, Osaka Women's and Children's Hospital, Osaka, Japan
| | | | | | | | - Masaya Kubota
- Division of Neurology, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
54
|
Abstract
The World Health Organization classifies diffuse low-grade gliomas (DLGGs) are highly epileptogenic primary brain tumors; epileptic seizures occur in more than 90% of cases. Epileptic seizures and drug resistance progress during the course of DLGGs. The glioma-related epileptogenic mechanisms are multifactorial; epileptogenic foci lie within the infiltrated peritumoral neocortex. A short seizure duration before surgery and a large extent of resection are the main predictors of postoperative seizure control in DLGGs. A supratotal resection of a DLGG can improve postoperative seizure control. Epileptic seizure at diagnosis positively affects DLGGs malignant transformation and overall survival.
Collapse
Affiliation(s)
- Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, 1 rue Cabanis, Paris Cedex 14 75674, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; French Glioma Study Group, Réseau d'Etude des Gliomes, REG, Groland, France; Inserm, U894, Centre Psychiatrie et Neurosciences, Paris, France.
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Medical Center, New York Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
55
|
Lalitha S, Minz RW, Medhi B. Understanding the controversial drug targets in epilepsy and pharmacoresistant epilepsy. Rev Neurosci 2018; 29:333-345. [PMID: 29211683 DOI: 10.1515/revneuro-2017-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/11/2017] [Indexed: 12/18/2022]
Abstract
Accumulating experimental data suggests a number of successful drug targets against epilepsy which eventually failed in the clinical setup. Mammalian target of rapamycin inhibitors, multi-drug resistance transporter inhibitors, cyclo-oxygenase-2 inhibitors, statins, etc. are the most promising and well studied among them. Drugs aiming at these targets produced beneficial response in most of the in vitro and in vivo seizure models. However, in certain situations, they have produced differential rather controversial results. Their effects varied with the seizure model, species, time and route of administration, different drugs from the same class, etc. This review emphasises on such drugs which presented with variability in their beneficial effects against seizures and epilepsy. This review critically summarises the preclinical evidence of these targets in the context of seizures and the probable reasons for their variability and clinical failures.
Collapse
Affiliation(s)
- Sree Lalitha
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ranjana W Minz
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
56
|
Abstract
Evidence from both preclinical and clinical studies suggest the importance of zinc homeostasis in seizures/epilepsy. Undoubtedly, zinc, via modulation of a variety of targets, is necessary for maintaining the balance between neuronal excitation and inhibition, while an imbalance between excitation and inhibition underlies seizures. However, the relationship between zinc signaling and seizures/epilepsy is complex as both extracellular and intracellular zinc may produce either protective or detrimental effects. This review provides an overview of preclinical/behavioral, functional and molecular studies, as well as clinical data on the involvement of zinc in the pathophysiology and treatment of seizures/epilepsy. Furthermore, the potential of targeting elements associated with zinc signaling or homeostasis and zinc levels as a therapeutic strategy for epilepsy is discussed.
Collapse
Affiliation(s)
- Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Katarzyna Młyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Aleksandra Wlaź
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland; Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
57
|
Hodges SL, Lugo JN. Wnt/β-catenin signaling as a potential target for novel epilepsy therapies. Epilepsy Res 2018; 146:9-16. [PMID: 30053675 DOI: 10.1016/j.eplepsyres.2018.07.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
Abstract
Epilepsy is one of the most common neurological disorders, and yet many afflicted individuals are resistant to all available therapeutic treatments. Existing pharmaceutical treatments function primarily to reduce hyperexcitability and prevent seizures, but fail to influence the underlying pathophysiology of the disorder. Recently, research efforts have focused on identifying alternative mechanistic targets for anti-epileptogenic therapies that can prevent the development of chronic epilepsy. The Wnt/β-catenin pathway, one possible target, has been demonstrated to be disrupted in both acute and chronic phases of epilepsy. Wnt/β-catenin signaling can regulate many seizure-induced changes in the brain, including neurogenesis and neuronal death, as well as can influence seizure susceptibility and potentially the development of chronic epilepsy. Several genome-wide studies and in vivo knockout animal models have provided evidence for an association between disrupted Wnt/β-catenin signaling and epilepsy. Furthermore, approved pharmaceutical drugs and other small molecule compounds that target components of the β-catenin destruction complex or antagonize endogenous inhibitors of the pathway have shown to be protective following seizures. However, additional studies are needed to determine the optimal time period in which modulation of the pathway may be most beneficial. Overall, disrupted molecular networks such as Wnt/β-catenin signaling, could be a promising anti-epileptogenic target for future epilepsy therapies.
Collapse
Affiliation(s)
- Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, TX, 76798, USA
| | - Joaquin N Lugo
- Institute of Biomedical Studies, Baylor University, Waco, TX, 76798, USA; Department of Psychology and Neuroscience, Baylor University, Waco, TX, 76798, USA; Department of Biology, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
58
|
Shirooie S, Nabavi SF, Dehpour AR, Belwal T, Habtemariam S, Argüelles S, Sureda A, Daglia M, Tomczyk M, Sobarzo-Sanchez E, Xu S, Nabavi SM. Targeting mTORs by omega-3 fatty acids: A possible novel therapeutic strategy for neurodegeneration? Pharmacol Res 2018; 135:37-48. [PMID: 29990625 DOI: 10.1016/j.phrs.2018.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases (NDs) such as Parkinson's (PD), Alzheimer's (AD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) cause significant world-wide morbidity and mortality. To date, there is no drug of cure for these, mostly age-related diseases, although approaches in delaying the pathology and/or giving patients some symptomatic relief have been adopted for the last few decades. Various studies in recent years have shown the beneficial effects of omega-3 poly unsaturated fatty acids (PUFAs) through diverse mechanisms including anti-inflammatory effects. This review now assesses the potential of this class of compounds in NDs therapy through specific action against the mammalian target of rapamycin (mTOR) signaling pathway. The role of mTOR in neurodegenerative diseases and targeted therapies by PUFAs are discussed.
Collapse
Affiliation(s)
- Samira Shirooie
- Department of Pharmacology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Fazel Nabavi
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 14359-16471, Iran
| | - Ahmad R Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tarun Belwal
- G.B. Pant National Institute of Himalayan Environment and Sustainable Development, Kosi Katarmal, Almora, Uttarakhand, India
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services UK, University of Greenwich, Chatham-Maritime, Kent ME4 4TB, UK
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, Italy
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
| | - Eduardo Sobarzo-Sanchez
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Spain; Instituto de Investigación en Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY 14623, United States
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 14359-16471, Iran.
| |
Collapse
|
59
|
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
|
60
|
Dynamic landscape of the local translation at activated synapses. Mol Psychiatry 2018; 23:107-114. [PMID: 29203851 PMCID: PMC5754473 DOI: 10.1038/mp.2017.245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 01/17/2023]
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is the central regulator of cap-dependent translation at the synapse. Disturbances in mTOR pathway have been associated with several neurological diseases, such as autism and epilepsy. RNA-binding protein FMRP, a negative regulator of translation initiation, is one of the key components of the local translation system. Activation and inactivation of FMRP occurs via phosphorylation by S6 kinase and dephosphorylation by PP2A phosphatase, respectively. S6 kinase and PP2A phosphatase are activated in response to mGluR receptor stimulation through different signaling pathways and at different rates. The dynamic aspects of this system are poorly understood. We developed a mathematical model of FMRP-dependent regulation of postsynaptic density (PSD) protein synthesis in response to mGluR receptor stimulation and conducted in silico experiments to study the regulatory circuit functioning. The modeling results revealed the possibility of generating oscillatory (cyclic and quasi-cyclic), chaotic and even hyperchaotic dynamics of postsynaptic protein synthesis as well as the presence of multiple attractors in a wide range of parameters of the local translation system. The results suggest that autistic disorders associated with mTOR pathway hyperactivation may be due to impaired proteome stability associated with the formation of complex dynamic regimes of PSD protein synthesis in response to stimulation of mGluR receptors on the postsynaptic membrane of excitatory synapses on pyramidal hippocampal cells.
Collapse
|
61
|
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
|
62
|
Lyu G, Han YL. [Research advances in hereditary epilepsy and precision drug therapy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:1118-1123. [PMID: 29046212 PMCID: PMC7389281 DOI: 10.7499/j.issn.1008-8830.2017.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/27/2017] [Indexed: 06/07/2023]
Abstract
Epilepsy is a common nervous system disease. It has been found that the pathogenesis of epilepsy is associated mutations in various genes, including genes encoding voltage-dependent ion channel, genes encoding ligand-gated ion channel, and solute carrier family genes. Different types of epilepsy caused by different mutations have different responses to drugs, and therefore, diagnosis and medication guidance based on genes are new thoughts for developing therapies. With the application of next-generation sequencing technology, more and more genes will be determined, which helps to further study the pathogenic mechanism of mutant genes and provides a basis for precision drug therapy for epilepsy.
Collapse
Affiliation(s)
- Ge Lyu
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning 530000, China.
| | | |
Collapse
|
63
|
Abstract
Advances in epilepsy treatment are occurring at a rapid pace, and it is challenging for us to keep up with the latest in our field. As we struggle to keep up with the literature and concentrate on our own research and clinical work, we often fail to exercise our imagination and envision what our field will be like in future decades. This was the assignment to the speakers for the Presidential Symposium at the 2016 American Epilepsy Society Annual Meeting. I challenged the experts to step outside the frame of their usual daily work to imagine what epilepsy treatment would and should look like for the next generation of epilepsy specialists and their patients. As you will read in the following sections, the speakers truly stepped up to the challenge to look into the crystal ball. The following are summaries of each lecture that describe the current state, existing cutting edge ideas, and some surprising predictions for the future. I am grateful for the tremendous effort these experts put into this challenge and hope they stimulate your imagination so you will work to bring these advances to our patients.
Collapse
|
64
|
Clossen BL, Reddy DS. Novel therapeutic approaches for disease-modification of epileptogenesis for curing epilepsy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1519-1538. [PMID: 28179120 PMCID: PMC5474195 DOI: 10.1016/j.bbadis.2017.02.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 11/16/2022]
Abstract
This article describes the recent advances in epileptogenesis and novel therapeutic approaches for the prevention of epilepsy, with a special emphasis on the pharmacological basis of disease-modification of epileptogenesis for curing epilepsy. Here we assess animal studies and human clinical trials of epilepsy spanning 1982-2016. Epilepsy arises from a number of neuronal factors that trigger epileptogenesis, which is the process by which a brain shifts from a normal physiologic state to an epileptic condition. The events precipitating these changes can be of diverse origin, including traumatic brain injury, cerebrovascular damage, infections, chemical neurotoxicity, and emergency seizure conditions such as status epilepticus. Expectedly, the molecular and system mechanisms responsible for epileptogenesis are not well defined or understood. To date, there is no approved therapy for the prevention of epilepsy. Epigenetic dysregulation, neuroinflammation, and neurodegeneration appear to trigger epileptogenesis. Targeted drugs are being identified that can truly prevent the development of epilepsy in at-risk people. The promising agents include rapamycin, COX-2 inhibitors, TRK inhibitors, epigenetic modulators, JAK-STAT inhibitors, and neurosteroids. Recent evidence suggests that neurosteroids may play a role in modulating epileptogenesis. A number of promising drugs are under investigation for the prevention or modification of epileptogenesis to halt the development of epilepsy. Some drugs in development appear rational for preventing epilepsy because they target the initial trigger or related signaling pathways as the brain becomes progressively more prone to seizures. Additional research into the target validity and clinical investigation is essential to make new frontiers in curing epilepsy.
Collapse
Affiliation(s)
- Bryan L Clossen
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
65
|
Blazejczyk M, Macias M, Korostynski M, Firkowska M, Piechota M, Skalecka A, Tempes A, Koscielny A, Urbanska M, Przewlocki R, Jaworski J. Kainic Acid Induces mTORC1-Dependent Expression of Elmo1 in Hippocampal Neurons. Mol Neurobiol 2017; 54:2562-2578. [PMID: 26993296 PMCID: PMC5390005 DOI: 10.1007/s12035-016-9821-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/29/2016] [Indexed: 12/24/2022]
Abstract
Epileptogenesis is a process triggered by initial environmental or genetic factors that result in epilepsy and may continue during disease progression. Important parts of this process include changes in transcriptome and the pathological rewiring of neuronal circuits that involves changes in neuronal morphology. Mammalian/mechanistic target of rapamycin (mTOR) is upregulated by proconvulsive drugs, e.g., kainic acid, and is needed for progression of epileptogenesis, but molecular aspects of its contribution are not fully understood. Since mTOR can modulate transcription, we tested if rapamycin, an mTOR complex 1 inhibitor, affects kainic acid-evoked transcriptome changes. Using microarray technology, we showed that rapamycin inhibits the kainic acid-induced expression of multiple functionally heterogeneous genes. We further focused on engulfment and cell motility 1 (Elmo1), which is a modulator of actin dynamics and therefore could contribute to pathological rewiring of neuronal circuits during epileptogenesis. We showed that prolonged overexpression of Elmo1 in cultured hippocampal neurons increased axonal growth, decreased dendritic spine density, and affected their shape. In conclusion, data presented herein show that increased mTORC1 activity in response to kainic acid has no global effect on gene expression. Instead, our findings suggest that mTORC1 inhibition may affect development of epilepsy, by modulating expression of specific subset of genes, including Elmo1, and point to a potential role for Elmo1 in morphological changes that accompany epileptogenesis.
Collapse
Affiliation(s)
- Magdalena Blazejczyk
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland.
| | - Matylda Macias
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Michal Korostynski
- Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Marcelina Firkowska
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Marcin Piechota
- Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Agnieszka Skalecka
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Aleksandra Tempes
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Alicja Koscielny
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Malgorzata Urbanska
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland
| | - Ryszard Przewlocki
- Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena St., 02-109, Warsaw, Poland.
| |
Collapse
|
66
|
Rossini L, Villani F, Granata T, Tassi L, Tringali G, Cardinale F, Aronica E, Spreafico R, Garbelli R. FCD Type II and mTOR pathway: Evidence for different mechanisms involved in the pathogenesis of dysmorphic neurons. Epilepsy Res 2016; 129:146-156. [PMID: 28056425 DOI: 10.1016/j.eplepsyres.2016.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/24/2016] [Accepted: 12/06/2016] [Indexed: 12/19/2022]
Abstract
Type II focal cortical dysplasia (FCD II) is a malformation of cortical development, frequently associated with intractable epilepsy, characterised by cortical dyslamination, dysmorphic neurons (DNs) and balloon cells (BCs). We investigated the expression of pS6 (downstream target) and pPDK1-pAkt (upstream targets) as evidence for mTOR pathway activation and their co-expression with Interleukin-1β in FCD II surgical specimens and compared the findings with control non-epileptic tissue, non-malformed epileptic tissue or acquired epilepsy-Rasmussen's Encephalitis (RE) occasionally presenting pS6 and Interleukin-1β positive abnormal neurons. Downstream mTOR activation was demonstrated in almost all abnormal cells in both FCD II and RE. Conversely, upstream activation in FCD II was observed in the majority of BCs, in a proportion of DNs, not presenting Interleukin-1β expression, but not at all in RE scattered abnormal neurons. Based on these findings we suggest that the presence of BCs and DNs in FCD II could be due to a first upstream mTOR pathway PI3K-Akt-mediate event occurring very early during cortical development in the large proportion of abnormal cells; followed by the appearance of additional pS6 positive DNs promoted by the presence of a later inflammatory processes.
Collapse
Affiliation(s)
- Laura Rossini
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy.
| | - Flavio Villani
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy
| | - Tiziana Granata
- Department of Pediatric Neuroscience, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy
| | - Laura Tassi
- Epilepsy Surgery Centre "C. Munari", Ospedale Niguarda, Milan, Italy
| | - Giovanni Tringali
- Neurosurgery Unit, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy
| | | | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience University of Amsterdam; Stichting Epilepsie Instellingen Nederland (SEIN), The Netherlands
| | - Roberto Spreafico
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy
| | - Rita Garbelli
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico "C. Besta", Milan, Italy
| |
Collapse
|
67
|
Liu JY, Reeves C, Diehl B, Coppola A, Al-Hajri A, Hoskote C, Mughairy SA, Tachrount M, Groves M, Michalak Z, Mills K, McEvoy AW, Miserocchi A, Sisodiya SM, Thom M. Early lipofuscin accumulation in frontal lobe epilepsy. Ann Neurol 2016; 80:882-895. [DOI: 10.1002/ana.24803] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Joan Y.W. Liu
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery; London United Kingdom
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
| | - Cheryl Reeves
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery; London United Kingdom
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
| | - Beate Diehl
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
- Department of Clinical Neurophysiology; National Hospital for Neurology and Neurosurgery; London United Kingdom
| | - Antonietta Coppola
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
| | - Aliya Al-Hajri
- The Lysholm Department of Neuroradiology in National Hospital for Neurology and Neurosurgery; London United Kingdom and Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - Chandrashekar Hoskote
- The Lysholm Department of Neuroradiology in National Hospital for Neurology and Neurosurgery; London United Kingdom and Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - Salim al Mughairy
- The Lysholm Department of Neuroradiology in National Hospital for Neurology and Neurosurgery; London United Kingdom and Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - Mohamed Tachrount
- The Lysholm Department of Neuroradiology in National Hospital for Neurology and Neurosurgery; London United Kingdom and Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - Michael Groves
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery; London United Kingdom
| | - Zuzanna Michalak
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery; London United Kingdom
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
| | - Kevin Mills
- Biological Mass Spectrometry Centre, Institute of Child Health; University College London; London United Kingdom
| | - Andrew W. McEvoy
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
- Victor Horsley Department of Neurosurgery; National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Anna Miserocchi
- Victor Horsley Department of Neurosurgery; National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Sanjay M. Sisodiya
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
- Epilepsy Society, Chesham Lane; Chalfont St Peter United Kingdom
| | - Maria Thom
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery; London United Kingdom
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London United Kingdom
| |
Collapse
|
68
|
Maiese K. Targeting molecules to medicine with mTOR, autophagy and neurodegenerative disorders. Br J Clin Pharmacol 2016; 82:1245-1266. [PMID: 26469771 PMCID: PMC5061806 DOI: 10.1111/bcp.12804] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 10/11/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are significantly increasing in incidence as the age of the global population continues to climb with improved life expectancy. At present, more than 30 million individuals throughout the world are impacted by acute and chronic neurodegenerative disorders with limited treatment strategies. The mechanistic target of rapamycin (mTOR), also known as the mammalian target of rapamycin, is a 289 kDa serine/threonine protein kinase that offers exciting possibilities for novel treatment strategies for a host of neurodegenerative diseases that include Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, stroke and trauma. mTOR governs the programmed cell death pathways of apoptosis and autophagy that can determine neuronal stem cell development, precursor cell differentiation, cell senescence, cell survival and ultimate cell fate. Coupled to the cellular biology of mTOR are a number of considerations for the development of novel treatments involving the fine control of mTOR signalling, tumourigenesis, complexity of the apoptosis and autophagy relationship, functional outcome in the nervous system, and the intimately linked pathways of growth factors, phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), AMP activated protein kinase (AMPK), silent mating type information regulation two homologue one (Saccharomyces cerevisiae) (SIRT1) and others. Effective clinical translation of the cellular signalling mechanisms of mTOR offers provocative avenues for new drug development in the nervous system tempered only by the need to elucidate further the intricacies of the mTOR pathway.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey, 07101, USA.
| |
Collapse
|
69
|
Sun Y, Lipton JO, Boyle LM, Madsen JR, Goldenberg MC, Pascual-Leone A, Sahin M, Rotenberg A. Direct current stimulation induces mGluR5-dependent neocortical plasticity. Ann Neurol 2016; 80:233-46. [PMID: 27315032 DOI: 10.1002/ana.24708] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To obtain insights into mechanisms mediating changes in cortical excitability induced by cathodal transcranial direct current stimulation (tDCS). METHODS Neocortical slices were exposed to direct current stimulation (DCS) delivered through Ag/AgCl electrodes over a range of current orientations, magnitudes, and durations. DCS-induced cortical plasticity and its receptor dependency were measured as the change in layer II/III field excitatory postsynaptic potentials by a multielectrode array, both with and without neurotransmitter receptor blockers or allosteric modulators. In vivo, tDCS was delivered to intact mice scalp via surface electrodes. Molecular consequences of DCS in vitro or tDCS in vivo were tested by immunoblot of protein extracted from stimulated slices or the neocortex harvested from stimulated intact mice. RESULTS Cathodal DCS in vitro induces a long-term depression (DCS-LTD) of excitatory synaptic strength in both human and mouse neocortical slices. DCS-LTD is abolished with an mGluR5 negative allosteric modulator, mechanistic target of rapamycin (mTOR) inhibitor, and inhibitor of protein synthesis. However, DCS-LTD persists despite either γ-aminobutyric acid type A receptor or N-methyl-D-aspartate receptor inhibition. An mGluR5-positive allosteric modulator, in contrast, transformed transient synaptic depression resultant from brief DCS application into durable DCS-LTD. INTERPRETATION We identify a novel molecular pathway by which tDCS modulates cortical excitability, and indicate a capacity for synergistic interaction between tDCS and pharmacologic mGluR5 facilitation. The findings support exploration of cathodal tDCS as a treatment of neurologic conditions characterized by aberrant regional cortical excitability referable to mGluR5-mTOR signaling. Ann Neurol 2016;80:233-246.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Neuromodulation Program, Boston Children's Hospital, Boston, MA.,Program in Neuroscience, Harvard Medical School, Boston, MA
| | - Jonathan O Lipton
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Lara M Boyle
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA
| | - Marti C Goldenberg
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Alexander Rotenberg
- Department of Neurology and F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Neuromodulation Program, Boston Children's Hospital, Boston, MA.,Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
70
|
The mTOR signalling cascade: paving new roads to cure neurological disease. Nat Rev Neurol 2016; 12:379-92. [PMID: 27340022 DOI: 10.1038/nrneurol.2016.81] [Citation(s) in RCA: 273] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Defining the multiple roles of the mechanistic (formerly 'mammalian') target of rapamycin (mTOR) signalling pathway in neurological diseases has been an exciting and rapidly evolving story of bench-to-bedside translational research that has spanned gene mutation discovery, functional experimental validation of mutations, pharmacological pathway manipulation, and clinical trials. Alterations in the dual contributions of mTOR - regulation of cell growth and proliferation, as well as autophagy and cell death - have been found in developmental brain malformations, epilepsy, autism and intellectual disability, hypoxic-ischaemic and traumatic brain injuries, brain tumours, and neurodegenerative disorders. mTOR integrates a variety of cues, such as growth factor levels, oxygen levels, and nutrient and energy availability, to regulate protein synthesis and cell growth. In line with the positioning of mTOR as a pivotal cell signalling node, altered mTOR activation has been associated with a group of phenotypically diverse neurological disorders. To understand how altered mTOR signalling leads to such divergent phenotypes, we need insight into the differential effects of enhanced or diminished mTOR activation, the developmental context of these changes, and the cell type affected by altered signalling. A particularly exciting feature of the tale of mTOR discovery is that pharmacological mTOR inhibitors have shown clinical benefits in some neurological disorders, such as tuberous sclerosis complex, and are being considered for clinical trials in epilepsy, autism, dementia, traumatic brain injury, and stroke.
Collapse
|
71
|
Verrotti A, Zara F, Minetti C, Striano P. Novel treatment perspectives from advances in understanding of genetic epilepsy syndromes. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1167594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
72
|
Citraro R, Leo A, Constanti A, Russo E, De Sarro G. mTOR pathway inhibition as a new therapeutic strategy in epilepsy and epileptogenesis. Pharmacol Res 2016; 107:333-343. [DOI: 10.1016/j.phrs.2016.03.039] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/23/2016] [Accepted: 03/31/2016] [Indexed: 12/24/2022]
|
73
|
Ye M, Bi YF, Ding L, Zhu WW, Gao W. Saikosaponin a functions as anti-epileptic effect in pentylenetetrazol induced rats through inhibiting mTOR signaling pathway. Biomed Pharmacother 2016; 81:281-287. [PMID: 27261605 DOI: 10.1016/j.biopha.2016.04.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE Saikosaponin a (SSa), which is one major bioactive compound isolated from radix bupleuri, has been demonstrated to exhibit the properties of anticonvulsant and antiepileptic in few reports. This study aims to clarify the molecular mechanism by which SSa protects against pentylenetetrazol (PTZ) induced epileptic seizure. METHODS PTZ induced rat and hippocampal neuron were established. Treated rats or hippocampal neuron with SSa, and mTOR, P70S6K, IL-1β and TNF-α were then determined. RESULTS In PTZ induced rat, SSa significantly reduced seizure severity and duration while markedly elevated seizure latency, and it also down-regulated hippocampal p-mTOR, p-70S6K, L-1β and TNF-α expression. In hippocampal neurons exposed to PTZ, p-mTOR and p-70S6K expression levels were also decreased by SSa. Pre-incubated hippocampal neurons with leucine, an mTOR agonist, reversed the effects of SSa on decreasing cytokines expression and inhibiting cell apoptosis. The treatment of mTOR inhibitor rapamycin prevented against the increase of cytokines expression and hippocampal neuron apoptosis induced by PTZ. Leucine also canceled the alleviation of seizures and induction of hippocampal caspase-3 activity in PTZ induced rat with the treatment of SSa. CONCLUSION SSa protects against PTZ induced epileptic seizure and hippocampal neuron apoptosis through inhibiting mTOR signaling pathway.
Collapse
Affiliation(s)
- Ming Ye
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Yong-Feng Bi
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Li Ding
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Wei-Wei Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow 215006, China
| | - Wei Gao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Soochow 215006, China.
| |
Collapse
|
74
|
Tuberous sclerosis--A model for tumour growth. Semin Cell Dev Biol 2016; 52:3-11. [PMID: 26816112 DOI: 10.1016/j.semcdb.2016.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/18/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder where patients develop benign tumours in several organ systems. Central to TSC pathology is hyper-activation of the mammalian target of rapamycin complex 1 (mTORC1) signalling pathway, which is a key controller of cell growth. As a result, TSC model systems are a valuable tool for examining mTORC1-driven cellular processes. The immunosuppressant, rapamycin, is a specific inhibitor of mTORC1 and has shown promise as a therapeutic agent in TSC as well as in malignancy. This review will focus on the cellular processes controlled by mTORC1 and how TSC-deficient cell lines and mouse models have broadened our understanding of the mTORC1 signalling network. It will also discuss how our knowledge of TSC signalling can help us understand sporadic conditions where mTORC1 activity is implicated in disease onset or progression, and the possibility of using rapamycin to treat sporadic disease.
Collapse
|
75
|
Ricos MG, Hodgson BL, Pippucci T, Saidin A, Ong YS, Heron SE, Licchetta L, Bisulli F, Bayly MA, Hughes J, Baldassari S, Palombo F, Santucci M, Meletti S, Berkovic SF, Rubboli G, Thomas PQ, Scheffer IE, Tinuper P, Geoghegan J, Schreiber AW, Dibbens LM. Mutations in the mammalian target of rapamycin pathway regulators NPRL2 and NPRL3 cause focal epilepsy. Ann Neurol 2015; 79:120-31. [PMID: 26505888 DOI: 10.1002/ana.24547] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/29/2015] [Accepted: 10/17/2015] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Focal epilepsies are the most common form observed and have not generally been considered to be genetic in origin. Recently, we identified mutations in DEPDC5 as a cause of familial focal epilepsy. In this study, we investigated whether mutations in the mammalian target of rapamycin (mTOR) regulators, NPRL2 and NPRL3, also contribute to cases of focal epilepsy. METHODS We used targeted capture and next-generation sequencing to analyze 404 unrelated probands with focal epilepsy. We performed exome sequencing on two families with multiple members affected with focal epilepsy and linkage analysis on one of these. RESULTS In our cohort of 404 unrelated focal epilepsy patients, we identified five mutations in NPRL2 and five in NPRL3. Exome sequencing analysis of two families with focal epilepsy identified NPRL2 and NPRL3 as the top candidate-causative genes. Some patients had focal epilepsy associated with brain malformations. We also identified 18 new mutations in DEPDC5. INTERPRETATION We have identified NPRL2 and NPRL3 as two new focal epilepsy genes that also play a role in the mTOR-signaling pathway. Our findings show that mutations in GATOR1 complex genes are the most significant cause of familial focal epilepsy identified to date, including cases with brain malformations. It is possible that deregulation of cellular growth control plays a more important role in epilepsy than is currently recognized.
Collapse
Affiliation(s)
- Michael G Ricos
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Bree L Hodgson
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Tommaso Pippucci
- Medical Genetics Unit, Polyclinic Sant'Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Akzam Saidin
- Novocraft Technologies Sdn Bhd, Selangor, Malaysia
| | - Yeh Sze Ong
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Sarah E Heron
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marta A Bayly
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - James Hughes
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Sara Baldassari
- Medical Genetics Unit, Polyclinic Sant'Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Flavia Palombo
- Medical Genetics Unit, Polyclinic Sant'Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Margherita Santucci
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefano Meletti
- Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, AUSL Modena, Modena, Italy
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Guido Rubboli
- Danish Epilepsy Center, Filadelfia/University of Copenhagen, Dianalund, Denmark.,IRCCS Institute of Neurological Sciences, Neurology Unit, Bellaria Hospital, Bologna, Italy
| | - Paul Q Thomas
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia.,Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Paolo Tinuper
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Joel Geoghegan
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Andreas W Schreiber
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,ACRF Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Leanne M Dibbens
- Epilepsy Research Program, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.,Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia.,Molecular Neurogenomics Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
76
|
Polypharmacology Shakes Hands with Complex Aetiopathology. Trends Pharmacol Sci 2015; 36:802-821. [PMID: 26434643 DOI: 10.1016/j.tips.2015.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/13/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023]
Abstract
Chronic diseases are due to deviations of fundamental physiological systems, with different pathologies being characterised by similar malfunctioning biological networks. The ensuing compensatory mechanisms may weaken the body's dynamic ability to respond to further insults and reduce the efficacy of conventional single target treatments. The multitarget, systemic, and prohomeostatic actions emerging for plant cannabinoids exemplify what might be needed for future medicines. Indeed, two combined cannabis extracts were approved as a single medicine (Sativex(®)), while pure cannabidiol, a multitarget cannabinoid, is emerging as a treatment for paediatric drug-resistant epilepsy. Using emerging cannabinoid medicines as an example, we revisit the concept of polypharmacology and describe a new empirical model, the 'therapeutic handshake', to predict efficacy/safety of compound combinations of either natural or synthetic origin.
Collapse
|
77
|
Wheless JW. Use of the mTOR inhibitor everolimus in a patient with multiple manifestations of tuberous sclerosis complex including epilepsy. EPILEPSY & BEHAVIOR CASE REPORTS 2015; 4:63-6. [PMID: 26543807 PMCID: PMC4543076 DOI: 10.1016/j.ebcr.2015.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 11/25/2022]
Abstract
Tuberous sclerosis complex (TSC) is a genetic disease in which overactivation of mechanistic target of rapamycin (mTOR) signaling leads to the growth of benign hamartomas in multiple organs, including the brain, and is associated with a high rate of epilepsy and neurological deficits. The mTOR inhibitor everolimus has been used in the treatment of subependymal giant cell astrocytomas and renal angiomyolipomas in patients with TSC. This article describes the case of a 13-year-old girl with TSC-associated epilepsy with refractory generalized seizures who initiated treatment with everolimus and experienced subsequent improvement in several TSC manifestations, including a reduction in seizure frequency from clusters of two or three daily to one every 2 to 4 weeks after 1.5 years of treatment.
Collapse
Affiliation(s)
- James W Wheless
- Le Bonheur Children's Hospital and the University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
78
|
Serrano E, Kanner AM. Recent treatment advances and novel therapeutic approaches in epilepsy. F1000PRIME REPORTS 2015; 7:61. [PMID: 26097734 PMCID: PMC4447056 DOI: 10.12703/p7-61] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The purpose of this article is to review recent advances in the treatment of epilepsy. It includes five antiepileptic drugs that have been recently added to the pharmacologic armamentarium and surgical techniques that have been developed in the last few years. Finally, we review ongoing research that may have a potential role in future treatments of epilepsy.
Collapse
|
79
|
Cambiaghi M, Magri L, Cursi M. Importance of EEG in validating the chronic effects of drugs: suggestions from animal models of epilepsy treated with rapamycin. Seizure 2015; 27:30-9. [PMID: 25891924 DOI: 10.1016/j.seizure.2015.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/12/2015] [Accepted: 02/15/2015] [Indexed: 11/29/2022] Open
Abstract
PURPOSE The development of new drugs for the treatment of epilepsy is a major challenge for modern neurology and its first steps demand basic research. Preclinical studies on animal models of epilepsy are mainly based on the analysis of brain electrical activity to detect seizures, when they are not just limited to behavioral tests like the Racine scale. METHODS In the present review, we discuss the importance of using time-locked video and EEG recordings (Video-EEG) coupled with behavioral tests as tools to monitor and analyze the effects of anti-epileptic drugs in pre-clinical research. Particularly, we focus on the utility of a multimodal approach based on EEG/behavioral analysis to study the beneficial effects of chronic rapamycin treatment as a potential anti-epileptogenic therapy for a broad spectrum of epilepsy, including both genetic (as in tuberous sclerosis complex) and acquired diseases. RESULTS Changes and synchronization of neuronal activity of different areas have been correlated with specific behavior in both physiological and pathological conditions. In the epileptic brain, during a seizure there is an abnormal activation of many cells all at once, altering different networks. CONCLUSION A multimodal approach based on video, EEG analysis and behavioral tests would be the best option in preclinical studies of epilepsy.
Collapse
Affiliation(s)
- Marco Cambiaghi
- Università degli Studi di Torino, Department of Neuroscience, Turin, Italy.
| | - Laura Magri
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Marco Cursi
- Clinical Neurophysiology Unit, Department of Neurology, Scientific Institute San Raffaele, Milan, Italy
| |
Collapse
|
80
|
Lin WH, He M, Baines RA. Seizure suppression through manipulating splicing of a voltage-gated sodium channel. ACTA ACUST UNITED AC 2015; 138:891-901. [PMID: 25681415 PMCID: PMC5014079 DOI: 10.1093/brain/awv012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Voltage-gated persistent sodium current (INaP) is a tractable target for antiepileptic drugs. Using a strategy focused on INaP reduction, Lin et al. identify 95 regulators of voltage-gated sodium channel splicing for which RNAi knockdown reduces seizure duration in Drosophila. Manipulation of splicing regulators could improve control of epilepsy. Seizure can result from increased voltage-gated persistent sodium current expression. Although many clinically-approved antiepileptic drugs target voltage-gated persistent sodium current, none exclusively repress this current without also adversely affecting the transient voltage-gated sodium current. Achieving a more selective block has significant potential for the treatment of epilepsy. Recent studies show that voltage-gated persistent sodium current amplitude is regulated by alternative splicing offering the possibility of a novel route for seizure control. In this study we identify 291 splicing regulators that, on knockdown, alter splicing of the Drosophila voltage-gated sodium channel to favour inclusion of exon K, rather than the mutually exclusive exon L. This change is associated with both a significant reduction in voltage-gated persistent sodium current, without change to transient voltage-gated sodium current, and to rescue of seizure in this model insect. RNA interference mediated knock-down, in two different seizure mutants, shows that 95 of these regulators are sufficient to significantly reduce seizure duration. Moreover, most suppress seizure activity in both mutants, indicative that they are part of well conserved pathways and likely, therefore, to be optimal candidates to take forward to mammalian studies. We provide proof-of-principle for such studies by showing that inhibition of a selection of regulators, using small molecule inhibitors, is similarly effective to reduce seizure. Splicing of the Drosophila sodium channel shows many similarities to its mammalian counterparts, including altering the amplitude of voltage-gated persistent sodium current. Our study provides the impetus to investigate whether manipulation of splicing of mammalian voltage-gated sodium channels may be exploitable to provide effective seizure control.
Collapse
Affiliation(s)
- Wei-Hsiang Lin
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Miaomiao He
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Richard A Baines
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
81
|
Radzik I, Miziak B, Dudka J, Chrościńska-Krawczyk M, Czuczwar SJ. Prospects of epileptogenesis prevention. Pharmacol Rep 2015; 67:663-8. [PMID: 25933984 DOI: 10.1016/j.pharep.2015.01.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 01/13/2023]
Abstract
Epilepsy is a common neurologic disease, affecting about 1-2% of the population. In around 30% of patients with epilepsy, their seizures are not satisfactorily controlled and drug-resistant epilepsy constitutes a real therapeutic challenge. Consequently, there are efforts aimed at the inhibition of epileptogenesis, a process of converting a normal into an epileptic brain. Data on this problem have been mainly obtained in post-status epilepticus rodent models in which spontaneous seizure activity and behavioral disturbances develop over time. Among antiepileptic drugs, diazepam at high dose of 20mg/kg given during status epilepticus, significantly inhibited the development of spontaneous seizures and also, a strong neuroprotective effect was evident. Also gabapentin and valproate (over a period of 40 days) proved effective in the inhibition of spontaneous seizure activity and reduction of behavioral deficit. However, there are also data that valproate (over 28 days) significantly improved the behavioral performance without affecting the occurrence of spontaneous seizures. A number of antiepileptic drugs, carbamazepine, lamotrigine, levetiracetam, phenobarbital, and topiramate were completely ineffective. Among non-antiepileptic drugs, some promise show rapamycin, losartan and combinations of anti-inflammatory drugs, targeting different inflammatory pathways. Inhibition of epileptogenesis may become a valuable therapeutic approach provided that there are reliable markers of this process. Actually, such markers begin to emerge.
Collapse
Affiliation(s)
- Iwona Radzik
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Medical University of Lublin, Lublin, Poland; Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Magdalena Chrościńska-Krawczyk
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Department of Pediatrics, Endocrinology and Neurology, Medical University of Lublin, Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Department of Physiopathology, Institute of Rural Health, Lublin, Poland.
| |
Collapse
|
82
|
Bejarano E, Rodríguez-Navarro JA. Autophagy and amino acid metabolism in the brain: implications for epilepsy. Amino Acids 2014; 47:2113-26. [DOI: 10.1007/s00726-014-1822-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/31/2014] [Indexed: 12/31/2022]
|
83
|
Evidence for mTOR pathway activation in a spectrum of epilepsy-associated pathologies. Acta Neuropathol Commun 2014; 2:71. [PMID: 25005575 PMCID: PMC4230418 DOI: 10.1186/2051-5960-2-71] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/06/2014] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Activation of the mTOR pathway has been linked to the cytopathology and epileptogenicity of malformations, specifically Focal Cortical Dysplasia (FCD) and Tuberous Sclerosis (TSC). Experimental and clinical trials have shown than mTOR inhibitors have anti-epileptogenic effects in TS. Dysmorphic neurones and balloon cells are hallmarks of FCDIIb and TSC, but similar cells are also occasionally observed in other acquired epileptogenic pathologies, including hippocampal sclerosis (HS) and Rasmussen's encephalitis (RE). Our aim was to explore mTOR pathway activation in a range of epilepsy-associated pathologies and in lesion-negative cases. RESULTS 50 epilepsy surgical pathologies were selected including HS ILAE type 1 with (5) and without dysmorphic neurones (4), FCDIIa (1), FCDIIb (5), FCDIIIa (5), FCDIIIb (3), FCDIIId (3), RE (5) and cortex adjacent to cavernoma (1). We also included pathology-negative epilepsy cases; temporal cortex (7), frontal cortex (2), paired frontal cortical samples with different ictal activity according to intracranial EEG recordings (4), cortex with acute injuries from electrode tracks (5) and additionally non-epilepsy surgical controls (3). Immunohistochemistry for phospho-S6 (pS6) ser240/244 and ser235/236 and double-labelling for Iba1, neurofilament, GFAP, GFAPdelta, doublecortin, and nestin were performed. Predominant neuronal labelling was observed with pS6 ser240/244 and glial labelling with pS6 ser235/236 in all pathology types but with evidence for co-expression in a proportion of cells in all pathologies. Intense labelling of dysmorphic neurones and balloon cells was observed in FCDIIb, but dysmorphic neurones were also labelled in RE and HS. There was no difference in pS6 labelling in paired samples according to ictal activity. Double-labelling immunofluorescent studies further demonstrated the co-localisation of pS6 with nestin, doublecortin, GFAPdelta in populations of small, immature neuroglial cells in a range of epilepsy pathologies. CONCLUSIONS Although mTOR activation has been more studied in the FCDIIb and TSC, our observations suggest this pathway is activated in a variety of epilepsy-associated pathologies, and in varied cell types including dysmorphic neurones, microglia and immature cell types. There was no definite evidence from our studies to suggest that pS6 expression is directly related to disease activity.
Collapse
|
84
|
Li MM, Jiang T, Sun Z, Zhang Q, Tan CC, Yu JT, Tan L. Genome-wide microRNA expression profiles in hippocampus of rats with chronic temporal lobe epilepsy. Sci Rep 2014; 4:4734. [PMID: 24751812 PMCID: PMC3994440 DOI: 10.1038/srep04734] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 04/01/2014] [Indexed: 12/18/2022] Open
Abstract
The expression and functions of microRNAs (miRNAs) in chronic temporal lobe epilepsy (TLE), the most common type of refractory epilepsy in adults, are poorly understood currently. In this study, status epilepticus evoked by amygdala stimulation was used to establish rat chronic TLE model. Two months later, high-throughput sequencing was employed to investigate miRNA expression profile in rat hippocampus, and six miRNAs were confirmed to be differentially expressed. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that most of the target genes for these six miRNAs were associated with neuronal apoptosis. Meanwhile, the levels of miR-423-3p and miR-296-5p were correlated with the activity of caspase-3, an apoptosis indicator. Additionally, the loading of miR-423-3p was increased in RNA-induced silencing complex whilst caspase-6, a target of miR-423-3p, was reduced in chronic TLE rats. Collectively, our findings suggest that miRNAs may exert anti-apoptotic effects in chronic TLE.
Collapse
Affiliation(s)
- Meng-Meng Li
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
- These authors contributed equally to this work
| | - Teng Jiang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
- These authors contributed equally to this work
| | - Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, Taishan Medical University, Qingdao, China
| | - Qun Zhang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Taishan Medical University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Taishan Medical University, Qingdao, China
| |
Collapse
|
85
|
Autophagy in aging and neurodegenerative diseases: implications for pathogenesis and therapy. Neurobiol Aging 2013; 35:941-57. [PMID: 24360503 DOI: 10.1016/j.neurobiolaging.2013.11.019] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/17/2013] [Accepted: 11/19/2013] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, share a common cellular and molecular pathogenetic mechanism involving aberrant misfolded protein or peptide aggregation and deposition. Autophagy represents a major route for degradation of aggregated cellular proteins and dysfunctional organelles. Emerging studies have demonstrated that up-regulation of autophagy can lead to decreased levels of these toxic aggregate-prone proteins, and is beneficial in the context of aging and various models of neurodegenerative diseases. Understanding the signaling pathways involved in the regulation of autophagy is crucial to the development of strategies for therapy. This review will discuss the cellular and molecular mechanisms of autophagy and its important role in the pathogenesis of aging and neurodegenerative diseases, and the ongoing drug discovery strategies for therapeutic modulation.
Collapse
|
86
|
Sun Z, Yu JT, Jiang T, Li MM, Tan L, Zhang Q, Tan L. Genome-wide microRNA profiling of rat hippocampus after status epilepticus induced by amygdala stimulation identifies modulators of neuronal apoptosis. PLoS One 2013; 8:e78375. [PMID: 24205215 PMCID: PMC3808371 DOI: 10.1371/journal.pone.0078375] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/20/2013] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are small and endogenously expressed non-coding RNAs that negatively regulate the expression of protein-coding genes at the translational level. Emerging evidence suggests that miRNAs play critical roles in central nervous system under physiological and pathological conditions. However, their expression and functions in status epilepticus (SE) have not been well characterized thus far. Here, by using high-throughput sequencing, we characterized miRNA expression profile in rat hippocampus at 24 hours following SE induced by amygdala stimulation. After confirmation by qRT-PCR, six miRNAs were found to be differentially expressed in brain after SE. Subsequent Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that most of the predicted target genes for these six miRNAs were related to neuronal apoptosis. We then investigated the dynamic changes of these six miRNAs at different time-point (4 hours, 24 hours, 1 week and 3 weeks) after SE. Meanwhile, neuronal survival and apoptosis in the hippocampus after SE were evaluated by Nissl staining and terminal deoxynucleotidyl transferase-mediated dUTP end-labeling assay. We found that the expression of miR-874-3p, miR-20a-5p, miR-345-3p, miR-365-5p, and miR-764-3p were significantly increased from 24 hours to 1 week, whereas miR-99b-3p level was markedly decreased from 24 hours to 3 weeks after SE. Further analysis revealed that the levels of miR-365-5p and miR-99b-3p were significantly correlated with neuronal apoptosis after SE. Taken together, our data suggest that miRNAs are important modulators of SE-induced neuronal apoptosis. These findings also open new avenues for future studies aimed at developing strategies against neuronal apoptosis after SE.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, Taishan Medical University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
- * E-mail: (Lan Tan); (J-TY)
| | - Teng Jiang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
| | - Meng-Meng Li
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Qun Zhang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Taishan Medical University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
- * E-mail: (Lan Tan); (J-TY)
| |
Collapse
|
87
|
Kaeberlein M. mTOR Inhibition: From Aging to Autism and Beyond. SCIENTIFICA 2013; 2013:849186. [PMID: 24379984 PMCID: PMC3860151 DOI: 10.1155/2013/849186] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/27/2013] [Indexed: 05/10/2023]
Abstract
The mechanistic target of rapamycin (mTOR) is a highly conserved protein that regulates growth and proliferation in response to environmental and hormonal cues. Broadly speaking, organisms are constantly faced with the challenge of interpreting their environment and making a decision between "grow or do not grow." mTOR is a major component of the network that makes this decision at the cellular level and, to some extent, the tissue and organismal level as well. Although overly simplistic, this framework can be useful when considering the myriad functions ascribed to mTOR and the pleiotropic phenotypes associated with genetic or pharmacological modulation of mTOR signaling. In this review, I will consider mTOR function in this context and attempt to summarize and interpret the growing body of literature demonstrating interesting and varied effects of mTOR inhibitors. These include robust effects on a multitude of age-related parameters and pathologies, as well as several other processes not obviously linked to aging or age-related disease.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, 1959 NE Pacific Street, D-514, Seattle, WA 98195-7470, USA
- *Matt Kaeberlein:
| |
Collapse
|