1
|
Bakouh N, Castaño-Martín R, Metais A, Dan EL, Balducci E, Chhuon C, Lepicka J, Barcia G, Losito E, Lourdel S, Planelles G, Muresan RC, Moca VV, Kaminska A, Bourgeois M, Chemaly N, Rguez Y, Auvin S, Huberfeld G, Varlet P, Asnafi V, Guerrera IC, Kabashi E, Nabbout R, Ciura S, Blauwblomme T. Chloride deregulation and GABA depolarization in MTOR-related malformations of cortical development. Brain 2025; 148:549-563. [PMID: 39106285 PMCID: PMC11788215 DOI: 10.1093/brain/awae262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/15/2024] [Accepted: 07/19/2024] [Indexed: 08/09/2024] Open
Abstract
Focal cortical dysplasia, hemimegalencephaly and cortical tubers are paediatric epileptogenic malformations of cortical development (MCDs) frequently pharmacoresistant and mostly treated surgically by the resection of epileptic cortex. Availability of cortical resection samples has allowed significant mechanistic discoveries directly from human material. Causal brain somatic or germline mutations in the AKT/PI3K/DEPDC5/MTOR genes have been identified. GABAA-mediated paradoxical depolarization, related to altered chloride (Cl-) homeostasis, has been shown to participate to ictogenesis in human paediatric MCDs. However, the link between genomic alterations and neuronal hyperexcitability is unclear. Here, we studied the post-translational interactions between the mTOR pathway and the regulation of cation-chloride cotransporters (CCCs), KCC2 and NKCC1, that are largely responsible for controlling intracellular Cl- and, ultimately, GABAergic transmission. For this study, 35 children (25 MTORopathies and 10 pseudo-controls, diagnosed by histology plus genetic profiling) were operated for drug-resistant epilepsy. Postoperative cortical tissues were recorded on a multi-electrode array to map epileptic activities. CCC expression level and phosphorylation status of the WNK1/SPAK-OSR1 pathway was measured during basal conditions and after pharmacological modulation. Direct interactions between mTOR and WNK1 pathway components were investigated by immunoprecipitation. Membranous incorporation of MCD samples in Xenopus laevis oocytes enabled measurement of the Cl- conductance and equilibrium potential for GABA. Of the 25 clinical cases, half harboured a somatic mutation in the mTOR pathway, and pS6 expression was increased in all MCD samples. Spontaneous interictal discharges were recorded in 65% of the slices. CCC expression was altered in MCDs, with a reduced KCC2/NKCC1 ratio and decreased KCC2 membranous expression. CCC expression was regulated by the WNK1/SPAK-OSR1 kinases through direct phosphorylation of Thr906 on KCC2, which was reversed by WNK1 and SPAK antagonists (N-ethylmaleimide and staurosporine). The mSIN1 subunit of MTORC2 was found to interact with SPAK-OSR1 and WNK1. Interactions between these key epileptogenic pathways could be reversed by the mTOR-specific antagonist rapamycin, leading to a dephosphorylation of CCCs and recovery of the KCC2/NKCC1 ratio. The functional effect of such recovery was validated by the restoration of the depolarizing shift in the equilibrium potential for GABA by rapamycin, measured after incorporation of MCD membranes into X. laevis oocytes, in line with a re-establishment of normal Cl- reversal potential. Our study deciphers a protein interaction network through a phosphorylation cascade between MTOR and WNK1/SPAK-OSR1 leading to deregulation of chloride cotransporters, increased neuronal Cl- levels and GABAA dysfunction in malformations of cortical development, linking genomic defects and functional effects and paving the way to target epilepsy therapy.
Collapse
Affiliation(s)
- Naziha Bakouh
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Reyes Castaño-Martín
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Alice Metais
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
- Service de Neuropathologie, GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | | | - Estelle Balducci
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Cerina Chhuon
- INSERM US24, Proteomic platform, SFR Necker, 75015 Paris, France
| | - Joanna Lepicka
- INSERM US24, Proteomic platform, SFR Necker, 75015 Paris, France
| | - Giulia Barcia
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Emma Losito
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Stéphane Lourdel
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, 75006 Paris, France
- CNRS EMR 8228—Laboratory of Renal Physiology and Tubulopathies, Université de Paris Cité, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Gabrielle Planelles
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, 75006 Paris, France
- CNRS EMR 8228—Laboratory of Renal Physiology and Tubulopathies, Université de Paris Cité, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Raul C Muresan
- STAR-UBB Institute, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Vasile Vlad Moca
- STAR-UBB Institute, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Anna Kaminska
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Marie Bourgeois
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Nicole Chemaly
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Yasmine Rguez
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
| | - Stéphane Auvin
- Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, 75019 Paris, France
| | - Gilles Huberfeld
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
| | - Pascale Varlet
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, 75014 Paris, France
- Service de Neuropathologie, GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | - Vahid Asnafi
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | | | - Edor Kabashi
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Rima Nabbout
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Sorana Ciura
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
| | - Thomas Blauwblomme
- Translational Research in Neuroscience Lab, Institut Imagine, Université Paris Cité, INSERM U1163, 75015 Paris, France
- Department of Pediatric Neurosurgery Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
- Department of Pediatric Neurology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| |
Collapse
|
2
|
Zhang J, Argueta D, Tong X, Vinters HV, Mathern GW, Cepeda C. Iconography of abnormal non-neuronal cells in pediatric focal cortical dysplasia type IIb and tuberous sclerosis complex. Front Cell Neurosci 2025; 18:1486315. [PMID: 39835291 PMCID: PMC11743721 DOI: 10.3389/fncel.2024.1486315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Once believed to be the culprits of epileptogenic activity, the functional properties of balloon/giant cells (BC/GC), commonly found in some malformations of cortical development including focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC), are beginning to be unraveled. These abnormal cells emerge during early brain development as a result of a hyperactive mTOR pathway and may express both neuronal and glial markers. A paradigm shift occurred when our group demonstrated that BC/GC in pediatric cases of FCDIIb and TSC are unable to generate action potentials and lack synaptic inputs. Hence, their role in epileptogenesis remained obscure. In this review, we provide a detailed characterization of abnormal non-neuronal cells including BC/GC, intermediate cells, and dysmorphic/reactive astrocytes found in FCDIIb and TSC cases, with special emphasis on electrophysiological and morphological assessments. Regardless of pathology, the electrophysiological properties of abnormal cells appear more glial-like, while others appear more neuronal-like. Their morphology also differs in terms of somatic size, shape, and dendritic elaboration. A common feature of these types of non-neuronal cells is their inability to generate action potentials. Thus, despite their distinct properties and etiologies, they share a common functional feature. We hypothesize that, although the exact role of abnormal non-neuronal cells in FCDIIb and TSC remains mysterious, it can be suggested that cells displaying more glial-like properties function in a similar way as astrocytes do, i.e., to buffer K+ ions and neurotransmitters, while those with more neuronal properties, may represent a metabolic burden due to high energy demands but inability to receive or transmit electric signals. In addition, due to the heterogeneity of these cells, a new classification scheme based on morphological, electrophysiological, and gene/protein expression in FCDIIb and TSC cases seems warranted.
Collapse
Affiliation(s)
- Joyce Zhang
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| | - Deneen Argueta
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| | - Xiaoping Tong
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, University of California - Los Angeles, Los Angeles, CA, United States
| | - Gary W. Mathern
- Department of Neurosurgery, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, United States
| | - Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California - Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
3
|
Cases‐Cunillera S, Quatraccioni A, Rossini L, Ruffolo G, Ono T, Baulac S, Auvin S, O'Brien TJ, Henshall DC, Akman Ö, Sankar R, Galanopoulou AS. WONOEP appraisal: The role of glial cells in focal malformations associated with early onset epilepsies. Epilepsia 2024; 65:3457-3468. [PMID: 39401070 PMCID: PMC11647439 DOI: 10.1111/epi.18126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024]
Abstract
Epilepsy represents a common neurological disorder in patients with developmental brain lesions, particularly in association with malformations of cortical development and low-grade glioneuronal tumors. In these diseases, genetic and molecular alterations in neurons are increasingly discovered that can trigger abnormalities in the neuronal network, leading to higher neuronal excitability levels. However, the mechanisms underlying epilepsy cannot rely solely on assessing the neuronal component. Growing evidence has revealed the high degree of complexity underlying epileptogenic processes, in which glial cells emerge as potential modulators of neuronal activity. Understanding the role of glial cells in developmental brain lesions such as malformations of cortical development and low-grade glioneuronal tumors is crucial due to the high degree of pharmacoresistance characteristic of these lesions. This has prompted research to investigate the role of glial and immune cells in epileptiform activity to find new therapeutic targets that could be used as combinatorial drug therapy. In a special session of the XVI Workshop of the Neurobiology of Epilepsy (WONOEP, Talloires, France, July 2022) organized by the Neurobiology Commission of the International League Against Epilepsy, we discussed the evidence exploring the genetic and molecular mechanisms of glial cells and immune response and their implications in the pathogenesis of neurodevelopmental pathologies associated with early life epilepsies.
Collapse
Affiliation(s)
- Silvia Cases‐Cunillera
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and GliomaParisFrance
| | - Anne Quatraccioni
- Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of BonnBonnGermany
| | - Laura Rossini
- Epilepsy UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Gabriele Ruffolo
- Department of Physiology and PharmacologyIstituto Pasteur–Fondazione Cenci Bolognetti, University of Rome SapienzaRomeItaly
- IRCCS San Raffaele RomaRomeItaly
| | - Tomonori Ono
- Epilepsy Center, National Hospital Organization Nagasaki Medical CenterŌmuraJapan
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, AP‐HP, Hôpital de la Pitié SalpêtrièreParisFrance
| | - Stéphane Auvin
- Pediatric Neurology Department, AP‐HP, Robert Debré University HospitalCRMR épilepsies Rares, EpiCARE memberParisFrance
- Université Paris Cité, INSERM NeuroDiderotParisFrance
- Institut Universitaire de FranceParisFrance
| | - Terence J. O'Brien
- Department of Neuroscience, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of NeurologyRoyal Melbourne HospitalMelbourneVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
- Department of Medicine (Royal Melbourne Hospital)University of MelbourneMelbourneVictoriaAustralia
| | - David C. Henshall
- Department of Physiology and Medical Physics, RCSIUniversity of Medicine and Health SciencesDublinIreland
| | - Özlem Akman
- Department of PhysiologyFaculty of Medicine, Demiroglu Bilim UniversityIstanbulTurkey
| | - Raman Sankar
- Department of Pediatrics and NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominique P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
4
|
Ellingson PJ, Shams YO, Parker JR, Calabrese RL, Cymbalyuk GS. Multistability of bursting rhythms in a half-center oscillator and the protective effects of synaptic inhibition. Front Cell Neurosci 2024; 18:1395026. [PMID: 39355175 PMCID: PMC11442309 DOI: 10.3389/fncel.2024.1395026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/08/2024] [Indexed: 10/03/2024] Open
Abstract
For animals to meet environmental challenges, the activity patterns of specialized oscillatory neural circuits, central pattern generators (CPGs), controlling rhythmic movements like breathing and locomotion, are adjusted by neuromodulation. As a representative example, the leech heartbeat is controlled by a CPG driven by two pairs of mutually inhibitory interneurons, heart interneuron (HN) half-center oscillators (HCO). Experiments and modeling indicate that neuromodulation of HCO navigates this CPG between dysfunctional regimes by employing a co-regulating inverted relation; reducing Na+/K+ pump current and increasing hyperpolarization-activated (h-) current. Simply reducing pump activity or increasing h-current leads to either seizure-like bursting or an asymmetric bursting dysfunctional regime, respectively. Here, we demonstrate through modeling that, alongside this coregulation path, a new bursting regime emerges. Both regimes fulfill the criteria for functional bursting activity. Although the cycle periods and burst durations of these patterns are roughly the same, the new one exhibits an intra-burst spike frequency that is twice as high as the other. This finding suggests that neuromodulation could introduce additional functional regimes with higher spike frequency, and thus more effective synaptic transmission to motor neurons. We found that this new regime co-exists with the original bursting. The HCO can be switched between them by a short pulse of excitatory or inhibitory conductance. In this domain of coexisting functional patterns, an isolated cell model exhibits only one regime, a severely dysfunctional plateau-containing, seizure-like activity. This aligns with widely reported notion that deficiency of inhibition can cause seizures and other dysfunctional neural activities. We show that along the coregulation path of neuromodulation, the high excitability of the single HNs induced by myomodulin is harnessed by mutually inhibitory synaptic interactions of the HCO into the functional bursting pattern.
Collapse
Affiliation(s)
- Parker J. Ellingson
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Yousif O. Shams
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Jessica R. Parker
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | | |
Collapse
|
5
|
Cho E, Kwon J, Lee G, Shin J, Lee H, Lee SH, Chung CK, Yoon J, Ho WK. Net synaptic drive of fast-spiking interneurons is inverted towards inhibition in human FCD I epilepsy. Nat Commun 2024; 15:6683. [PMID: 39107293 PMCID: PMC11303528 DOI: 10.1038/s41467-024-51065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Focal cortical dysplasia type I (FCD I) is the most common cause of pharmaco-resistant epilepsy with the poorest prognosis. To understand the epileptogenic mechanisms of FCD I, we obtained tissue resected from patients with FCD I epilepsy, and from tumor patients as control. Using whole-cell patch clamp in acute human brain slices, we investigated the cellular properties of fast-spiking interneurons (FSINs) and pyramidal neurons (PNs) within the ictal onset zone. In FCD I epilepsy, FSINs exhibited lower firing rates from slower repolarization and action potential broadening, while PNs had increased firing. Importantly, excitatory synaptic drive of FSINs increased progressively with the scale of cortical activation as a general property across species, but this relationship was inverted towards net inhibition in FCD I epilepsy. Further comparison with intracranial electroencephalography (iEEG) from the same patients revealed that the spatial extent of pathological high-frequency oscillations (pHFO) was associated with synaptic events at FSINs.
Collapse
Affiliation(s)
- Eunhye Cho
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Jii Kwon
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Gyuwon Lee
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Jiwoo Shin
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Hyunsu Lee
- Department of Physiology, Pusan National University School of Medicine, Busan, Korea
| | - Suk-Ho Lee
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea
| | - Chun Kee Chung
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea.
- Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, Korea.
| | - Jaeyoung Yoon
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Won-Kyung Ho
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea.
| |
Collapse
|
6
|
Ribierre T, Bacq A, Donneger F, Doladilhe M, Maletic M, Roussel D, Le Roux I, Chassoux F, Devaux B, Adle-Biassette H, Ferrand-Sorbets S, Dorfmüller G, Chipaux M, Baldassari S, Poncer JC, Baulac S. Targeting pathological cells with senolytic drugs reduces seizures in neurodevelopmental mTOR-related epilepsy. Nat Neurosci 2024; 27:1125-1136. [PMID: 38710875 PMCID: PMC11156583 DOI: 10.1038/s41593-024-01634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/28/2024] [Indexed: 05/08/2024]
Abstract
Cortical malformations such as focal cortical dysplasia type II (FCDII) are associated with pediatric drug-resistant epilepsy that necessitates neurosurgery. FCDII results from somatic mosaicism due to post-zygotic mutations in genes of the PI3K-AKT-mTOR pathway, which produce a subset of dysmorphic cells clustered within healthy brain tissue. Here we show a correlation between epileptiform activity in acute cortical slices obtained from human surgical FCDII brain tissues and the density of dysmorphic neurons. We uncovered multiple signatures of cellular senescence in these pathological cells, including p53/p16 expression, SASP expression and senescence-associated β-galactosidase activity. We also show that administration of senolytic drugs (dasatinib/quercetin) decreases the load of senescent cells and reduces seizure frequency in an MtorS2215F FCDII preclinical mouse model, providing proof of concept that senotherapy may be a useful approach to control seizures. These findings pave the way for therapeutic strategies selectively targeting mutated senescent cells in FCDII brain tissue.
Collapse
Affiliation(s)
- Théo Ribierre
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- NeuroNA Human Cellular Neuroscience Platform, Fondation Campus Biotech Geneva, Geneva, Switzerland
| | - Alexandre Bacq
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Florian Donneger
- Institut du Fer à Moulin, INSERM, Sorbonne Université, UMR-S 1270, Paris, France
| | - Marion Doladilhe
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marina Maletic
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Delphine Roussel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Isabelle Le Roux
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Francine Chassoux
- Service de Neurochirurgie, AP-HP, Hôpital Lariboisière, Paris, France
- GHU Paris, Psychiatrie et Neurosciences, Paris, France
| | - Bertrand Devaux
- Service de Neurochirurgie, AP-HP, Hôpital Lariboisière, Paris, France
- GHU Paris, Psychiatrie et Neurosciences, Paris, France
| | - Homa Adle-Biassette
- Université de Paris Cité, Service d'Anatomie Pathologique, AP-HP, Hôpital Lariboisière, DMU DREAM, UMR 1141, INSERM, Paris, France
| | | | - Georg Dorfmüller
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, Paris, France
| | - Mathilde Chipaux
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, Paris, France
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | | | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France.
| |
Collapse
|
7
|
Wise DL, Greene SB, Escobedo-Lozoya Y, Van Hooser SD, Nelson SB. Progressive Circuit Hyperexcitability in Mouse Neocortical Slice Cultures with Increasing Duration of Activity Silencing. eNeuro 2024; 11:ENEURO.0362-23.2024. [PMID: 38653560 PMCID: PMC11079856 DOI: 10.1523/eneuro.0362-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Forebrain neurons deprived of activity become hyperactive when activity is restored. Rebound activity has been linked to spontaneous seizures in vivo following prolonged activity blockade. Here, we measured the time course of rebound activity and the contributing circuit mechanisms using calcium imaging, synaptic staining, and whole-cell patch clamp in organotypic slice cultures of mouse neocortex. Calcium imaging revealed hypersynchronous activity increasing in intensity with longer periods of deprivation. While activity partially recovered 3 d after slices were released from 5 d of deprivation, they were less able to recover after 10 d of deprivation. However, even after the longer period of deprivation, activity patterns eventually returned to baseline levels. The degree of deprivation-induced rebound was age-dependent, with the greatest effects occurring when silencing began in the second week. Pharmacological blockade of NMDA receptors indicated that hypersynchronous rebound activity did not require activation of Hebbian plasticity. In single-neuron recordings, input resistance roughly doubled with a concomitant increase in intrinsic excitability. Synaptic imaging of pre- and postsynaptic proteins revealed dramatic reductions in the number of presumptive synapses with a larger effect on inhibitory than excitatory synapses. Putative excitatory synapses colocalizing PSD-95 and Bassoon declined by 39 and 56% following 5 and 10 d of deprivation, but presumptive inhibitory synapses colocalizing gephyrin and VGAT declined by 55 and 73%, respectively. The results suggest that with prolonged deprivation, a progressive reduction in synapse number is accompanied by a shift in the balance between excitation and inhibition and increased cellular excitability.
Collapse
Affiliation(s)
- Derek L Wise
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | - Samuel B Greene
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | | | | | - Sacha B Nelson
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| |
Collapse
|
8
|
Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes MTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. eLife 2024; 12:RP91010. [PMID: 38411613 PMCID: PMC10942629 DOI: 10.7554/elife.91010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activator genes, Rheb or MTOR, or biallelic inactivation of the mTORC1 repressor genes, Depdc5, Tsc1, or Pten in the mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
Collapse
Affiliation(s)
- Lena H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at DallasRichardsonUnited States
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Youfen Xu
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Maanasi Nair
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
9
|
Zheng Y, Xu C, Sun J, Ming W, Dai S, Shao Y, Qiu X, Li M, Shen C, Xu J, Fei F, Fang J, Jiang X, Zheng G, Hu W, Wang Y, Wang S, Ding M, Chen Z. Excitatory somatostatin interneurons in the dentate gyrus drive a widespread seizure network in cortical dysplasia. Signal Transduct Target Ther 2023; 8:186. [PMID: 37193687 DOI: 10.1038/s41392-023-01404-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/19/2023] [Accepted: 03/05/2023] [Indexed: 05/18/2023] Open
Abstract
Seizures due to cortical dysplasia are notorious for their poor prognosis even with medications and surgery, likely due to the widespread seizure network. Previous studies have primarily focused on the disruption of dysplastic lesions, rather than remote regions such as the hippocampus. Here, we first quantified the epileptogenicity of the hippocampus in patients with late-stage cortical dysplasia. We further investigated the cellular substrates leading to the epileptic hippocampus, using multiscale tools including calcium imaging, optogenetics, immunohistochemistry and electrophysiology. For the first time, we revealed the role of hippocampal somatostatin-positive interneurons in cortical dysplasia-related seizures. Somatostatin-positive were recruited during cortical dysplasia-related seizures. Interestingly, optogenetic studies suggested that somatostatin-positive interneurons paradoxically facilitated seizure generalization. By contrast, parvalbumin-positive interneurons retained an inhibitory role as in controls. Electrophysiological recordings and immunohistochemical studies revealed glutamate-mediated excitatory transmission from somatostatin-positive interneurons in the dentate gyrus. Taken together, our study reveals a novel role of excitatory somatostatin-positive neurons in the seizure network and brings new insights into the cellular basis of cortical dysplasia.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Cenglin Xu
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Jinyi Sun
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenjie Ming
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Sijie Dai
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuying Shao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Menghan Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chunhong Shen
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jinghong Xu
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Fan Fei
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiajia Fang
- Department of Neurology, Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Xuhong Jiang
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Guoqing Zheng
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
| | - Weiwei Hu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi Wang
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Shuang Wang
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Meiping Ding
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Zhong Chen
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
10
|
Righes Marafiga J, Baraban SC. Cell therapy for neurological disorders: Progress towards an embryonic medial ganglionic eminence progenitor-based treatment. Front Neurosci 2023; 17:1177678. [PMID: 37123353 PMCID: PMC10140420 DOI: 10.3389/fnins.2023.1177678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Impairment of development, migration, or function of inhibitory interneurons are key features of numerous circuit-based neurological disorders, such as epilepsy. From a therapeutic perspective, symptomatic treatment of these disorders often relies upon drugs or deep brain stimulation approaches to provide a general enhancement of GABA-mediated inhibition. A more effective strategy to target these pathological circuits and potentially provide true disease-modifying therapy, would be to selectively add new inhibitory interneurons into these circuits. One such strategy, using embryonic medial ganglionic (MGE) progenitor cells as a source of a unique sub-population of interneurons, has already proven effective as a cell transplantation therapy in a variety of preclinical models of neurological disorders, especially in mouse models of acquired epilepsy. Here we will discuss the evolution of this interneuron-based transplantation therapy in acquired epilepsy models, with an emphasis on the recent adaptation of MGE progenitor cells for xenotransplantation into larger mammals.
Collapse
Affiliation(s)
- Joseane Righes Marafiga
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Scott C. Baraban
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
- Helen Wills Institute for Neuroscience, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
11
|
Weir JS, Christiansen N, Sandvig A, Sandvig I. Selective inhibition of excitatory synaptic transmission alters the emergent bursting dynamics of in vitro neural networks. Front Neural Circuits 2023; 17:1020487. [PMID: 36874945 PMCID: PMC9978115 DOI: 10.3389/fncir.2023.1020487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Neurons in vitro connect to each other and form neural networks that display emergent electrophysiological activity. This activity begins as spontaneous uncorrelated firing in the early phase of development, and as functional excitatory and inhibitory synapses mature, the activity typically emerges as spontaneous network bursts. Network bursts are events of coordinated global activation among many neurons interspersed with periods of silencing and are important for synaptic plasticity, neural information processing, and network computation. While bursting is the consequence of balanced excitatory-inhibitory (E/I) interactions, the functional mechanisms underlying their evolution from physiological to potentially pathophysiological states, such as decreasing or increasing in synchrony, are still poorly understood. Synaptic activity, especially that related to maturity of E/I synaptic transmission, is known to strongly influence these processes. In this study, we used selective chemogenetic inhibition to target and disrupt excitatory synaptic transmission in in vitro neural networks to study functional response and recovery of spontaneous network bursts over time. We found that over time, inhibition resulted in increases in both network burstiness and synchrony. Our results indicate that the disruption in excitatory synaptic transmission during early network development likely affected inhibitory synaptic maturity which resulted in an overall decrease in network inhibition at later stages. These findings lend support to the importance of E/I balance in maintaining physiological bursting dynamics and, conceivably, information processing capacity in neural networks.
Collapse
Affiliation(s)
- Janelle Shari Weir
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nicholas Christiansen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Neurology and Clinical Neurophysiology, St. Olav's University Hospital, Trondheim, Norway.,Division of Neuro, Head and Neck, Department of Pharmacology and Clinical Neurosciences, Umeå University Hospital, Umeå, Sweden.,Division of Neuro, Head and Neck, Department of Community Medicine and Rehabilitation, Umeå University Hospital, Umeå, Sweden
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
12
|
Shakhatreh L, Janmohamed M, Baker AA, Willard A, Laing J, Rychkova M, Chen Z, Kwan P, O'Brien TJ, Perucca P. Interictal and seizure-onset EEG patterns in malformations of cortical development: A systematic review. Neurobiol Dis 2022; 174:105863. [PMID: 36165814 DOI: 10.1016/j.nbd.2022.105863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/07/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Malformations of cortical development (MCDs) are common causes of drug-resistant epilepsy. The mechanisms underlying the associated epileptogenesis and ictogenesis remain poorly elucidated. EEG can help in understanding these mechanisms. We systematically reviewed studies reporting scalp or intracranial EEG features of MCDs to characterise interictal and seizure-onset EEG patterns across different MCD types. METHODS We conducted a systematic review in accordance with PRISMA guidelines. MEDLINE, PubMed, and Cochrane databases were searched for studies describing interictal and seizure-onset EEG patterns in MCD patients. A classification framework was implemented to group EEG features into 20 predefined patterns, comprising nine interictal (five, scalp EEG; four, intracranial EEG) and 11 seizure-onset (five, scalp EEG; six, intracranial EEG) patterns. Logistic regression was used to estimate the odds ratios (OR) of each seizure-onset pattern being associated with specific MCD types. RESULTS Our search yielded 1682 studies, of which 27 comprising 936 MCD patients were included. Of the nine interictal EEG patterns, five (three, scalp EEG; two, intracranial EEG) were detected in ≥2 MCD types, while four (rhythmic epileptiform discharges type 1 and type 2 on scalp EEG; repetitive bursting spikes and sporadic spikes on intracranial EEG) were seen only in focal cortical dysplasia (FCD). Of the 11 seizure-onset patterns, eight (three, scalp EEG; five, intracranial EEG) were found in ≥2 MCD types, whereas three were observed only in FCD (suppression on scalp EEG; delta brush on intracranial EEG) or tuberous sclerosis complex (TSC; focal fast wave on scalp EEG). Among scalp EEG seizure-onset patterns, paroxysmal fast activity (OR = 0.13; 95% CI: 0.03-0.53; p = 0.024) and repetitive epileptiform discharges (OR = 0.18; 95% CI: 0.05-0.61; p = 0.036) were less likely to occur in TSC than FCD. Among intracranial EEG seizure-onset patterns, low-voltage fast activity was more likely to be detected in heterotopia (OR = 19.3; 95% CI: 6.22-60.1; p < 0.001), polymicrogyria (OR = 6.70; 95% CI: 2.25-20.0; p = 0.004) and TSC (OR = 4.27; 95% CI: 1.88-9.70; p = 0.005) than FCD. SIGNIFICANCE Different MCD types can share similar interictal or seizure-onset EEG patterns, reflecting common underlying biological mechanisms. However, selected EEG patterns appear to point to distinct MCD types, suggesting certain differences in their neuronal networks.
Collapse
Affiliation(s)
- Lubna Shakhatreh
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia.
| | - Mubeen Janmohamed
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia
| | - Ana Antonic Baker
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia
| | - Anna Willard
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia
| | - Joshua Laing
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia
| | - Maria Rychkova
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia
| | - Zhibin Chen
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia
| | - Patrick Kwan
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia; Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia; Department of Medicine (The Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Piero Perucca
- Department of Neuroscience, The Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia; Department of Neurology, Alfred Health, Melbourne, Australia; Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Melbourne, Australia; Epilepsy Research Centre, Department of Medicine (Austin Health), The University of Melbourne, Melbourne, Australia
| |
Collapse
|
13
|
Shao Y, Ge Q, Yang J, Wang M, Zhou Y, Guo JX, Zhu M, Shi J, Hu Y, Shen L, Chen Z, Li XM, Zhu JM, Zhang J, Duan S, Chen J. Pathological Networks Involving Dysmorphic Neurons in Type II Focal Cortical Dysplasia. Neurosci Bull 2022; 38:1007-1024. [PMID: 35235180 PMCID: PMC9468210 DOI: 10.1007/s12264-022-00828-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/24/2021] [Indexed: 10/19/2022] Open
Abstract
Focal cortical dysplasia (FCD) is one of the most common causes of drug-resistant epilepsy. Dysmorphic neurons are the major histopathological feature of type II FCD, but their role in seizure genesis in FCD is unclear. Here we performed whole-cell patch-clamp recording and morphological reconstruction of cortical principal neurons in postsurgical brain tissue from drug-resistant epilepsy patients. Quantitative analyses revealed distinct morphological and electrophysiological characteristics of the upper layer dysmorphic neurons in type II FCD, including an enlarged soma, aberrant dendritic arbors, increased current injection for rheobase action potential firing, and reduced action potential firing frequency. Intriguingly, the upper layer dysmorphic neurons received decreased glutamatergic and increased GABAergic synaptic inputs that were coupled with upregulation of the Na+-K+-Cl- cotransporter. In addition, we found a depolarizing shift of the GABA reversal potential in the CamKII-cre::PTENflox/flox mouse model of drug-resistant epilepsy, suggesting that enhanced GABAergic inputs might depolarize dysmorphic neurons. Thus, imbalance of synaptic excitation and inhibition of dysmorphic neurons may contribute to seizure genesis in type II FCD.
Collapse
Affiliation(s)
- Yijie Shao
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Department of Neurosurgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qianqian Ge
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jiachao Yang
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mi Wang
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yu Zhou
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jin-Xin Guo
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Mengyue Zhu
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jiachen Shi
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yiqi Hu
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li Shen
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Department of Orthopedic Surgery, School of Medicine, the Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, China
- Hangzhou Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310058, China
| | - Xiao-Ming Li
- Center for Neuroscience and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Center for Brain Science and Brain-Inspired Intelligence, Joint Institute for Genetics and Genome Medicine between, Guangdong Hong Kong Macao Greater Bay Area, Zhejiang University and the University of Toronto, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun-Ming Zhu
- Department of Neurosurgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Shumin Duan
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Jiadong Chen
- Center for Neuroscience and Department of Neurosurgery of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
14
|
Fasano G, Compagnucci C, Dallapiccola B, Tartaglia M, Lauri A. Teleost Fish and Organoids: Alternative Windows Into the Development of Healthy and Diseased Brains. Front Mol Neurosci 2022; 15:855786. [PMID: 36034498 PMCID: PMC9403253 DOI: 10.3389/fnmol.2022.855786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The variety in the display of animals’ cognition, emotions, and behaviors, typical of humans, has its roots within the anterior-most part of the brain: the forebrain, giving rise to the neocortex in mammals. Our understanding of cellular and molecular events instructing the development of this domain and its multiple adaptations within the vertebrate lineage has progressed in the last decade. Expanding and detailing the available knowledge on regionalization, progenitors’ behavior and functional sophistication of the forebrain derivatives is also key to generating informative models to improve our characterization of heterogeneous and mechanistically unexplored cortical malformations. Classical and emerging mammalian models are irreplaceable to accurately elucidate mechanisms of stem cells expansion and impairments of cortex development. Nevertheless, alternative systems, allowing a considerable reduction of the burden associated with animal experimentation, are gaining popularity to dissect basic strategies of neural stem cells biology and morphogenesis in health and disease and to speed up preclinical drug testing. Teleost vertebrates such as zebrafish, showing conserved core programs of forebrain development, together with patients-derived in vitro 2D and 3D models, recapitulating more accurately human neurogenesis, are now accepted within translational workflows spanning from genetic analysis to functional investigation. Here, we review the current knowledge of common and divergent mechanisms shaping the forebrain in vertebrates, and causing cortical malformations in humans. We next address the utility, benefits and limitations of whole-brain/organism-based fish models or neuronal ensembles in vitro for translational research to unravel key genes and pathological mechanisms involved in neurodevelopmental diseases.
Collapse
|
15
|
Gomes I, Jesus Ribeiro J, Palavra F. Monitoring and Managing Patients with Tuberous Sclerosis Complex: Current State of Knowledge. J Multidiscip Healthc 2022; 15:1469-1480. [PMID: 35860622 PMCID: PMC9292455 DOI: 10.2147/jmdh.s266990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/29/2022] [Indexed: 12/05/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disease of autosomal dominant transmission that, in most cases, results from the presence of pathogenic variants of the TSC1 or TSC2 genes, encoding hamartin and tuberin, respectively. It is a multisystemic disease, affecting most frequently the brain, skin, kidney, and heart. The wide variety of possible clinical manifestations, given this multisystem dimension, makes the follow-up of patients with TSC an exercise of multidisciplinarity. In fact, these patients may require the intervention of various medical specialties, which thus have to combine their efforts to practice a medicine that is truly holistic. The past few years have witnessed a dramatic leap not only in the diagnosis and management of TSC patients, with standard monitoring recommendations, but also in the therapeutic field, with the use of mTORC1 inhibitors. In this article, we review the clinical manifestations associated with TSC, as well as the treatment and follow-up strategies that should be implemented, from a multidisciplinary perspective.
Collapse
Affiliation(s)
- Inês Gomes
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Filipe Palavra
- Center for Child Development - Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
16
|
Investigating the Role of GABA in Neural Development and Disease Using Mice Lacking GAD67 or VGAT Genes. Int J Mol Sci 2022; 23:ijms23147965. [PMID: 35887307 PMCID: PMC9318753 DOI: 10.3390/ijms23147965] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
Normal development and function of the central nervous system involves a balance between excitatory and inhibitory neurotransmission. Activity of both excitatory and inhibitory neurons is modulated by inhibitory signalling of the GABAergic and glycinergic systems. Mechanisms that regulate formation, maturation, refinement, and maintenance of inhibitory synapses are established in early life. Deviations from ideal excitatory and inhibitory balance, such as down-regulated inhibition, are linked with many neurological diseases, including epilepsy, schizophrenia, anxiety, and autism spectrum disorders. In the mammalian forebrain, GABA is the primary inhibitory neurotransmitter, binding to GABA receptors, opening chloride channels and hyperpolarizing the cell. We review the involvement of down-regulated inhibitory signalling in neurological disorders, possible mechanisms for disease progression, and targets for therapeutic intervention. We conclude that transgenic models of disrupted inhibitory signalling—in GAD67+/− and VGAT−/− mice—are useful for investigating the effects of down-regulated inhibitory signalling in a range of neurological diseases.
Collapse
|
17
|
Reorganization of Parvalbumin Immunopositive Perisomatic Innervation of Principal Cells in Focal Cortical Dysplasia Type IIB in Human Epileptic Patients. Int J Mol Sci 2022; 23:ijms23094746. [PMID: 35563137 PMCID: PMC9100614 DOI: 10.3390/ijms23094746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
Focal cortical dysplasia (FCD) is one of the most common causes of drug-resistant epilepsy. As several studies have revealed, the abnormal functioning of the perisomatic inhibitory system may play a role in the onset of seizures. Therefore, we wanted to investigate whether changes of perisomatic inhibitory inputs are present in FCD. Thus, the input properties of abnormal giant- and control-like principal cells were examined in FCD type IIB patients. Surgical samples were compared to controls from the same cortical regions with short postmortem intervals. For the study, six subjects were selected/each group. The perisomatic inhibitory terminals were quantified in parvalbumin and neuronal nuclei double immunostained sections using a confocal fluorescent microscope. The perisomatic input of giant neurons was extremely abundant, whereas control-like cells of the same samples had sparse inputs. A comparison of pooled data shows that the number of parvalbumin-immunopositive perisomatic terminals contacting principal cells was significantly larger in epileptic cases. The analysis showed some heterogeneity among epileptic samples. However, five out of six cases had significantly increased perisomatic input. Parameters of the control cells were homogenous. The reorganization of the perisomatic inhibitory system may increase the probability of seizure activity and might be a general mechanism of abnormal network activity.
Collapse
|
18
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
19
|
Yang T, Hu S, Chang WC, Kao HY, Wang Y. Perineuronal Nets Degradation and Parvalbumin Interneuron Loss in a Mouse Model of DEPDC5-Related Epilepsy. Dev Neurosci 2022; 44:671-677. [PMID: 35580549 PMCID: PMC9669280 DOI: 10.1159/000525039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
DEPDC5, the key gene within the mechanistic target of rapamycin (mTOR) pathway, is one of the most common causative genes in patients with epilepsy and malformation of cortical development (MCD). Although somatic mutations in the dorsal cortical progenitors generate the malformed cortex, its pathogenesis of hyperexcitability is complex and remains unclear. We specifically deleted Depdc5 in the mouse forebrain dorsal progenitors to model DEPDC5-related epilepsy and investigated whether and how parvalbumin interneurons were non-cell autonomously affected in the malformed cortex. We showed that long before seizures, coincident with microglia inflammation, proteolytic enzymes degraded perineuronal nets (PNNs) in the malformed cortex, resulting in parvalbumin (PV+) interneuron loss and presynaptic inhibition impairment. Our studies, therefore, uncovered the hitherto unknown role of PNN in mTOR-related MCD, providing a new framework for mechanistic-based therapeutic development.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Shuntong Hu
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Wei-Chih Chang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Hsin-Yi Kao
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| |
Collapse
|
20
|
Cheng L, Xing Y, Zhang H, Liu R, Lai H, Piao Y, Wang W, Yan X, Li X, Wang J, Li D, Loh HH, Yu T, Zhang G, Yang X. Mechanistic Analysis of Micro-Neurocircuits Underlying the Epileptogenic Zone in Focal Cortical Dysplasia Patients. Cereb Cortex 2021; 32:2216-2230. [PMID: 34664065 DOI: 10.1093/cercor/bhab350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
We aim to explore the microscopic neurophysiology of focal cortical dysplasia (FCD) induced epileptogenesis in specific macroscopic brain regions, therefore mapping a micro-macro neuronal network that potentially indicates the epileptogenic mechanism. Epileptic and relatively non-epileptic temporal neocortex specimens were resected from FCD and mesial temporal lobe epilepsy (mTLE) patients, respectively. Whole-cell patch-clamping was performed on cells from the seizure onset zone (SOZ) and non-SOZ inside the epileptogenic zone (EZ) of FCD patients, as well as the non-epileptic neocortex of mTLE patients. Microscopic data were recorded, including membrane characteristics, spontaneous synaptic activities, and evoked action potentials. Immunohistochemistry was also performed on parvalbumin-positive (PV+) interneurons. We found that SOZ interneurons exhibited abnormal neuronal expression and distribution as well as reduced overall function compared with non-SOZ and mTLE interneurons. The SOZ pyramidal cells experienced higher excitation but lower inhibition than the mTLE controls, whereas the non-SOZ pyramidal cells exhibited intermediate excitability. Action potential properties of both types of neurons also suggested more synchronized neuronal activity inside the EZ, particularly inside the SOZ. Together, our research provides evidence for a potential neurocircuit underlying SOZ epileptogenesis and non-SOZ seizure susceptibility. Further investigation of this microscopic network may promote understanding of the mechanism of FCD-induced epileptogenesis.
Collapse
Affiliation(s)
- Lipeng Cheng
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yue Xing
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Herui Zhang
- Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China
| | - Ru Liu
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Huanling Lai
- Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China
| | - Yueshan Piao
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wei Wang
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoming Yan
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaonan Li
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiaoyang Wang
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Donghong Li
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangdong 510635, China
| | - Horace H Loh
- Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China
| | - Tao Yu
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Guojun Zhang
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.,Functional Neurosurgery Department, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Xiaofeng Yang
- Center of Epilepsy, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.,Fundamental Research Department, Guangzhou Laboratory, Guangzhou 510700, China.,Neuroelectrophysiological Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
21
|
Berg J, Sorensen SA, Ting JT, Miller JA, Chartrand T, Buchin A, Bakken TE, Budzillo A, Dee N, Ding SL, Gouwens NW, Hodge RD, Kalmbach B, Lee C, Lee BR, Alfiler L, Baker K, Barkan E, Beller A, Berry K, Bertagnolli D, Bickley K, Bomben J, Braun T, Brouner K, Casper T, Chong P, Crichton K, Dalley R, de Frates R, Desta T, Lee SD, D'Orazi F, Dotson N, Egdorf T, Enstrom R, Farrell C, Feng D, Fong O, Furdan S, Galakhova AA, Gamlin C, Gary A, Glandon A, Goldy J, Gorham M, Goriounova NA, Gratiy S, Graybuck L, Gu H, Hadley K, Hansen N, Heistek TS, Henry AM, Heyer DB, Hill D, Hill C, Hupp M, Jarsky T, Kebede S, Keene L, Kim L, Kim MH, Kroll M, Latimer C, Levi BP, Link KE, Mallory M, Mann R, Marshall D, Maxwell M, McGraw M, McMillen D, Melief E, Mertens EJ, Mezei L, Mihut N, Mok S, Molnar G, Mukora A, Ng L, Ngo K, Nicovich PR, Nyhus J, Olah G, Oldre A, Omstead V, Ozsvar A, Park D, Peng H, Pham T, Pom CA, Potekhina L, Rajanbabu R, Ransford S, Reid D, Rimorin C, Ruiz A, Sandman D, Sulc J, Sunkin SM, Szafer A, Szemenyei V, Thomsen ER, Tieu M, Torkelson A, Trinh J, Tung H, Wakeman W, Waleboer F, Ward K, Wilbers R, Williams G, Yao Z, Yoon JG, Anastassiou C, Arkhipov A, Barzo P, Bernard A, Cobbs C, de Witt Hamer PC, Ellenbogen RG, Esposito L, Ferreira M, Gwinn RP, Hawrylycz MJ, Hof PR, Idema S, Jones AR, Keene CD, Ko AL, Murphy GJ, Ng L, Ojemann JG, Patel AP, Phillips JW, Silbergeld DL, Smith K, Tasic B, Yuste R, Segev I, de Kock CPJ, Mansvelder HD, Tamas G, Zeng H, Koch C, Lein ES. Human neocortical expansion involves glutamatergic neuron diversification. Nature 2021; 598:151-158. [PMID: 34616067 PMCID: PMC8494638 DOI: 10.1038/s41586-021-03813-8] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/07/2021] [Indexed: 11/09/2022]
Abstract
The neocortex is disproportionately expanded in human compared with mouse1,2, both in its total volume relative to subcortical structures and in the proportion occupied by supragranular layers composed of neurons that selectively make connections within the neocortex and with other telencephalic structures. Single-cell transcriptomic analyses of human and mouse neocortex show an increased diversity of glutamatergic neuron types in supragranular layers in human neocortex and pronounced gradients as a function of cortical depth3. Here, to probe the functional and anatomical correlates of this transcriptomic diversity, we developed a robust platform combining patch clamp recording, biocytin staining and single-cell RNA-sequencing (Patch-seq) to examine neurosurgically resected human tissues. We demonstrate a strong correspondence between morphological, physiological and transcriptomic phenotypes of five human glutamatergic supragranular neuron types. These were enriched in but not restricted to layers, with one type varying continuously in all phenotypes across layers 2 and 3. The deep portion of layer 3 contained highly distinctive cell types, two of which express a neurofilament protein that labels long-range projection neurons in primates that are selectively depleted in Alzheimer's disease4,5. Together, these results demonstrate the explanatory power of transcriptomic cell-type classification, provide a structural underpinning for increased complexity of cortical function in humans, and implicate discrete transcriptomic neuron types as selectively vulnerable in disease.
Collapse
Affiliation(s)
- Jim Berg
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Eliza Barkan
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Allison Beller
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kyla Berry
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Kris Bickley
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Peter Chong
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Tsega Desta
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - David Feng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Olivia Fong
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Szabina Furdan
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Anna A Galakhova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Clare Gamlin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Natalia A Goriounova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | - Hong Gu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Tim S Heistek
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Alex M Henry
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Djai B Heyer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - DiJon Hill
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Chris Hill
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Madie Hupp
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Jarsky
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Sara Kebede
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lisa Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Lisa Kim
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Caitlin Latimer
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Rusty Mann
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Desiree Marshall
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Medea McGraw
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Erica Melief
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Eline J Mertens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Leona Mezei
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Norbert Mihut
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | | | - Gabor Molnar
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Alice Mukora
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lindsay Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Gaspar Olah
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Aaron Oldre
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Attila Ozsvar
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Daniel Park
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | - David Reid
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Aaron Szafer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Viktor Szemenyei
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | | | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Femke Waleboer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Katelyn Ward
- Allen Institute for Brain Science, Seattle, WA, USA
| | - René Wilbers
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Pal Barzo
- Department of Neurosurgery, University of Szeged, Szeged, Hungary
| | - Amy Bernard
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Philip C de Witt Hamer
- Cancer Center Amsterdam, Brain Tumor Center, Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | | | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sander Idema
- Cancer Center Amsterdam, Brain Tumor Center, Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Andrew L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Gabe J Murphy
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeffrey G Ojemann
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Anoop P Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | - Daniel L Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | | | - Rafael Yuste
- NeuroTechnology Center, Columbia University, New York, NY, USA
| | - Idan Segev
- Edmond and Lily Safra Center for Brain Sciences and Department of Neurobiology, The Hebrew University Jerusalem, Jerusalem, Israel
| | - Christiaan P J de Kock
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, The Netherlands
| | - Gabor Tamas
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged, Hungary
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA.
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Ste20-like Kinase Is Critical for Inhibitory Synapse Maintenance and Its Deficiency Confers a Developmental Dendritopathy. J Neurosci 2021; 41:8111-8125. [PMID: 34400520 DOI: 10.1523/jneurosci.0352-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/18/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
The size and structure of the dendritic arbor play important roles in determining how synaptic inputs of neurons are converted to action potential output. The regulatory mechanisms governing the development of dendrites, however, are insufficiently understood. The evolutionary conserved Ste20/Hippo kinase pathway has been proposed to play an important role in regulating the formation and maintenance of dendritic architecture. A key element of this pathway, Ste20-like kinase (SLK), regulates cytoskeletal dynamics in non-neuronal cells and is strongly expressed throughout neuronal development. However, its function in neurons is unknown. We show that, during development of mouse cortical neurons, SLK has a surprisingly specific role for proper elaboration of higher, ≥ third-order dendrites both in male and in female mice. Moreover, we demonstrate that SLK is required to maintain excitation-inhibition balance. Specifically, SLK knockdown caused a selective loss of inhibitory synapses and functional inhibition after postnatal day 15, whereas excitatory neurotransmission was unaffected. Finally, we show that this mechanism may be relevant for human disease, as dysmorphic neurons within human cortical malformations revealed significant loss of SLK expression. Overall, the present data identify SLK as a key regulator of both dendritic complexity during development and inhibitory synapse maintenance.SIGNIFICANCE STATEMENT We show that dysmorphic neurons of human epileptogenic brain lesions have decreased levels of the Ste20-like kinase (SLK). Decreasing SLK expression in mouse neurons revealed that SLK has essential functions in forming the neuronal dendritic tree and in maintaining inhibitory connections with neighboring neurons.
Collapse
|
23
|
Righes Marafiga J, Vendramin Pasquetti M, Calcagnotto ME. GABAergic interneurons in epilepsy: More than a simple change in inhibition. Epilepsy Behav 2021; 121:106935. [PMID: 32035792 DOI: 10.1016/j.yebeh.2020.106935] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/20/2022]
Abstract
The pathophysiology of epilepsy has been historically grounded on hyperexcitability attributed to the oversimplified imbalance between excitation (E) and inhibition (I) in the brain. The decreased inhibition is mostly attributed to deficits in gamma-aminobutyric acid-containing (GABAergic) interneurons, the main source of inhibition in the central nervous system. However, the cell diversity, the wide range of spatiotemporal connectivity, and the distinct effects of the neurotransmitter GABA especially during development, must be considered to critically revisit the concept of hyperexcitability caused by decreased inhibition as a key characteristic in the development of epilepsy. Here, we will discuss that behind this known mechanism, there is a heterogeneity of GABAergic interneurons with distinct functions and sources, which have specific roles in controlling the neural network activity within the recruited microcircuit and altered network during the epileptogenic process. This article is part of the Special Issue "NEWroscience 2018.
Collapse
Affiliation(s)
- Joseane Righes Marafiga
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Mayara Vendramin Pasquetti
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Biological Science: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre 90046-900, RS, Brazil.
| |
Collapse
|
24
|
Koh HY, Jang J, Ju SH, Kim R, Cho GB, Kim DS, Sohn JW, Paik SB, Lee JH. Non-Cell Autonomous Epileptogenesis in Focal Cortical Dysplasia. Ann Neurol 2021; 90:285-299. [PMID: 34180075 DOI: 10.1002/ana.26149] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Low-level somatic mosaicism in the brain has been shown to be a major genetic cause of intractable focal epilepsy. However, how a relatively few mutation-carrying neurons are able to induce epileptogenesis at the local network level remains poorly understood. METHODS To probe the origin of epileptogenesis, we measured the excitability of neurons with MTOR mutation and nearby nonmutated neurons recorded by whole-cell patch-clamp and array-based electrodes comparing the topographic distribution of mutation. Computational simulation is used to understand neural network-level changes based on electrophysiological properties. To examine the underlying mechanism, we measured inhibitory and excitatory synaptic inputs in mutated neurons and nearby neurons by electrophysiological and histological methods using the mouse model and postoperative human brain tissue for cortical dysplasia. To explain non-cell-autonomous hyperexcitability, an inhibitor of adenosine kinase was injected into mice to enhance adenosine signaling and to mitigate hyperactivity of nearby nonmutated neurons. RESULTS We generated mice with a low-level somatic mutation in MTOR presenting spontaneous seizures. The seizure-triggering hyperexcitability originated from nonmutated neurons near mutation-carrying neurons, which proved to be less excitable than nonmutated neurons. Interestingly, the net balance between excitatory and inhibitory synaptic inputs onto mutated neurons remained unchanged. Additionally, we found that inhibition of adenosine kinase, which affects adenosine metabolism and neuronal excitability, reduced the hyperexcitability of nonmutated neurons. INTERPRETATION This study shows that neurons carrying somatic mutations in MTOR lead to focal epileptogenesis via non-cell-autonomous hyperexcitability of nearby nonmutated neurons. ANN NEUROL 2021;90:285-299.
Collapse
Affiliation(s)
- Hyun Yong Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jaeson Jang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sang Hyeon Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ryunhee Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Gyu-Bon Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Dong Seok Kim
- Department of Neurosurgery, Pediatric Epilepsy Clinics, Brain Korea 21 Project for Medical Science, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.,Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.,SoVarGen, Daejeon, Republic of Korea
| |
Collapse
|
25
|
Gong T, Liu Y, Chen Y, Lin L, Lin Y, Wang G. Focal corticarl dysplasia in epilepsy is associated with GABA increase. Neuroimage Clin 2021; 31:102763. [PMID: 34280836 PMCID: PMC8313738 DOI: 10.1016/j.nicl.2021.102763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/01/2022]
Abstract
PURPOSE Focal cortical dysplasia (FCD) is a major cause of drug-resistant epilepsy; however the underlying epileptogenic mechanisms of FCD metabolism in epilepsy patients remain unclear. The aim of this study is to detect alterations of γ-aminobutyric acid (GABA), glutathione (GSH), and the composite of glutamate and glutamine (Glx) in MRI-typical and neuropathologically confirmed FCD-associated epilepsy using Hadamard Encoding and Reconstruction of Mega-Edited Spectroscopy (HERMES). MATERIALS AND METHODS Fourteen epileptic patients suspected to be caused by FCD and 14 healthy controls were enrolled prospectively in this study; all subjects underwent a 3 T MRI scan, including 3D T1 weighted imaging and HERMES. The GABA signal detected by HERMES also contains signals from macromolecules and homocarnosine, so it is referred as GABA+. Signals of GABA+, GSH and Glx detected by HERMES from tumor foci, contralateral cerebral regions, and healthy controls were quantified using Gannet. Fitting errors and signal to noise ratios (SNRs) of GABA + signals were also recorded. Differences of GABA+, GSH, Glx, fitting error and SNR of GABA + among three groups were analyzed using linear mixed effects models. RESULTS Twelve FCD-associated epilepsy patients (7 females, aged 21.9 ± 9.3 years) and 12 matched healthy controls (7 females, aged 22.8 ± 9.8 years) were finally enrolled in this study. ANOVA results indicated that GABA levels were significantly increased in FCD foci compared with contralateral regions (p = 0.008) and with healthy controls (p = 0.003), while no difference was found in GSH and Glx levels. No difference of fitting errors or SNR of GABA + was found among FCD foci, contralateral regions and healthy controls. CONCLUSIONS Increased GABA levels were found in FCD foci that indicated GABA may play a central role in the pathophysiology of FCD patients with epilepsy.
Collapse
Affiliation(s)
- Tao Gong
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; MSunSoft Health Group, Shandong, China
| | - Yubo Liu
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yufan Chen
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | | | - Youting Lin
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Guangbin Wang
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China.
| |
Collapse
|
26
|
Ragot A, Luhmann HJ, Dipper-Wawra M, Heinemann U, Holtkamp M, Fidzinski P. Pathology-selective antiepileptic effects in the focal freeze-lesion rat model of malformation of cortical development. Exp Neurol 2021; 343:113776. [PMID: 34058228 DOI: 10.1016/j.expneurol.2021.113776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/13/2021] [Accepted: 05/27/2021] [Indexed: 01/29/2023]
Abstract
Malformations of cortical development (MCD) represent a group of rare diseases with severe clinical presentation as epileptic and pharmacoresistant encephalopathies. Morphological studies in tissue from MCD patients have revealed reduced GABAergic efficacy and increased intracellular chloride concentration in neuronal cells as important pathophysiological mechanisms in MCD. Also, in various animal models, alterations of GABAergic inhibition have been postulated as a predominant factor contributing to perilesional hyperexcitability. Along with this line, the NKCC1 inhibitor bumetanide has been postulated as a potential drug for treatment of epilepsy, mediating its antiepileptic effect by reduction of the intracellular chloride and increased inhibitory efficacy of GABAergic transmission. In the present study, we focused on the focal freeze-lesion model of MCD to compare antiepileptic drugs with distinct mechanisms of action, including NKCC1 inhibition by bumetanide. For this purpose, we combined electrophysiological and optical methods in slice preparations and assessed the properties of seizure like events (SLE) induced by 4-aminopyridine. In freeze-lesioned but not control slices, SLE onset was confined to the perilesional area, confirming that this region is hyperexcitable and likely triggers pathological activity. Bumetanide selectively reduced epileptic activity in lesion-containing slices but not in slices from sham-treated control rats. Moreover, bumetanide caused a shift in the SLE onset site away from the perilesional area. In contrast, effects of other antiepileptic drugs including carbamazepine, lacosamide, acezatolamide and zonisamide occurred mostly independently of the lesion and did not result in a shift of the onset region. Our work adds evidence for the functional relevance of chloride homeostasis in the pathophysiology of microgyrus formation as represented in the focal freeze-lesion model. Further studies in different MCD models and human tissue will be required to validate the effects across different MCD subtypes and species and to assess the translational value of our findings.
Collapse
Affiliation(s)
- Aliénor Ragot
- Charité - Universitätsmedizin Berlin, Clinical and Experimental Epileptology, Department of Neurology, Charitéplatz 1, 10117 Berlin, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Dipper-Wawra
- Charité - Universitätsmedizin Berlin, Clinical and Experimental Epileptology, Department of Neurology, Charitéplatz 1, 10117 Berlin, Germany
| | - Uwe Heinemann
- Charité - Universitätsmedizin Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Martin Holtkamp
- Charité - Universitätsmedizin Berlin, Clinical and Experimental Epileptology, Department of Neurology, Charitéplatz 1, 10117 Berlin, Germany; Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany
| | - Pawel Fidzinski
- Charité - Universitätsmedizin Berlin, Clinical and Experimental Epileptology, Department of Neurology, Charitéplatz 1, 10117 Berlin, Germany; Epilepsy-Center Berlin-Brandenburg, Institute for Diagnostics of Epilepsy, Berlin, Germany; Berlin Institute of Health at Charité -Universitätsmedizin Berlin, NeuroCure Clinical Research Center, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
27
|
Lazzarotto G, Klippel Zanona Q, Cagliari Zenki K, Calcagnotto ME. Effect of Memantine on Pentylenetetrazol-induced Seizures and EEG Profile in Animal Model of Cortical Malformation. Neuroscience 2021; 457:114-124. [PMID: 33465407 DOI: 10.1016/j.neuroscience.2020.12.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/27/2020] [Accepted: 12/31/2020] [Indexed: 11/28/2022]
Abstract
Developmental cortical malformations (DCM) are one of the main causes of refractory epilepsy. Many are the mechanisms underlying the hyperexcitability in DCM, including the important contribution of N-methyl-D-aspartate receptors (NMDAR). NMDAR blockers are shown to abolish seizures and epileptiform activity. Memantine, a NMDAR antagonist used to treat Alzheimeŕs disease, has been recently investigated as a possible treatment for other neurological disorders. However, the effects on preventing or diminishing seizures are controversial. Here we aimed to evaluate the effects of memantine on pentylenetetrazole (PTZ)-induced seizures in the freeze-lesion (FL) model. Bilateral cortical microgyria were induced (FL) or not (Sham) in male Wistar neonate rats. At P30, subdural electrodes were implanted and 7 days later, video-EEG was recorded in animals receiving either memantine (FL-M or Sham-M) or saline (FL-S or Sham-S), followed by PTZ. Seizures were evaluated by video-EEG during one hour and scored according to Racine scale. The video-EEG analyses revealed that the number of seizures and the total duration of stage IV-V seizures developed during the 1 h-period increased after memantine application in all groups. The EEG power spectral density (PSD) analysis showed an increased PSD of pre-ictal delta in Sham-M animals and increased PSD of slow, middle and fast gamma oscillations after memantine injection that persists during the pre-ictal period in all groups. Our findings suggested that memantine was unable to control the PTZ-induced seizures and that the associated enhancement of PSD of gamma oscillations may contribute to the increased probability of seizure development in these animals.
Collapse
Affiliation(s)
- Gabriela Lazzarotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Querusche Klippel Zanona
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Kamila Cagliari Zenki
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory (NNNESP Lab.), Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
28
|
Banerjee J, Dey S, Dixit AB, Doddamani R, Sharma MC, Garg A, Chandra PS, Tripathi M. GABA A Receptor-Mediated Epileptogenicity in Focal Cortical Dysplasia (FCD) Depends on Age at Epilepsy Onset. Front Cell Neurosci 2020; 14:562811. [PMID: 33192309 PMCID: PMC7556289 DOI: 10.3389/fncel.2020.562811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/01/2020] [Indexed: 11/13/2022] Open
Abstract
Enhanced spontaneous GABAA receptor activity is associated with focal cortical dysplasia (FCD), a developmental malformation of the cerebral cortex. Clinical manifestations in FCD vary with age at epilepsy onset with a more favorable prognosis in patients with late-onset (LO) compared to that in cases with early-onset (EO). This study was designed to test the hypothesis in FCD that spontaneous GABAA receptor-mediated epileptogenicity depends on the age at epilepsy onset and varies between patients with early and late-onset age in FCD. To this end, brain specimens were obtained from the maximal spiking region (MAX) and minimal spiking region (MIN) of the epileptic foci of EO (n = 14, mean age = 10.6 ± 2.9 years) and LO (n = 10, mean age = 27 ± 5.6 years) patients undergoing electrocorticography (ECoG) guided surgery. The whole-cell patch-clamp technique was used to record spontaneous GABAergic currents from normal-looking pyramidal neurons in slice preparations of resected brain samples. We detected higher frequency and amplitude of GABAergic events in MAX samples compared to MIN samples of LO patients, while they were comparable in MIN and MAX samples of EO patients. Further GABAergic activity in the MIN and MAX samples of EO patients was higher than the MIN samples of LO patients. This suggests that in LO patients, GABAA receptor-mediated epileptogenicity is confined only to the high spiking areas, but in EO patients, it affects low spiking regions as well.
Collapse
Affiliation(s)
- Jyotirmoy Banerjee
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Soumil Dey
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Aparna Banerjee Dixit
- Dr. B R Ambedkar Centre for Biomedical Research, University of Delhi, New Delhi, India
| | - Ramesh Doddamani
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Meher Chand Sharma
- Department of Neuropathology, All India Institute of Medical Sciences, New Delhi, India
| | - Ajay Garg
- Department of Neuroimaging and Interventional Neuroradiology, All India Institute of Medical Sciences, New Delhi, India
| | - P Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
29
|
GABAergic Interneuron and Neurotransmission Are mTOR-Dependently Disturbed in Experimental Focal Cortical Dysplasia. Mol Neurobiol 2020; 58:156-169. [PMID: 32909150 DOI: 10.1007/s12035-020-02086-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/21/2020] [Indexed: 01/13/2023]
Abstract
Focal cortical dysplasia (FCD) is a major cause for drug-resistant epilepsies. The molecular and cellular mechanisms of epileptogenesis in FCD are still poorly understood. Some studies have suggested that deficiencies of γ-aminobutyric acid (GABA) system may play an important role in type II FCD, but it remains controversial. In order to examine whether and how GABAergic interneurons and synaptic function are affected, we generated a somatic mTOR hyperactivation-based mouse model of type II FCD by in utero electroporation, quantified densities of interneurons in the malformed cortices, and recorded miniature inhibitory postsynaptic currents in dysmorphic neurons. We detected 20-25% reduction of GABAergic interneurons within malformed cortices, independent of cortical regions and cell subtypes but proportionate to the decrease of global neuron counts. GABAergic synaptic transmission from interneurons to mTOR hyperactivated dysmorphic neurons was dramatically disrupted, outweighing the decrease of interneuron counts. Postnatal mTOR inhibition partially rescued these alterations of GABAergic system. We also quantified the expression of GABAA receptor, GABA transporter, and chloridion transporter encoding genes and found that their expression was relatively intact within the malformed cortices. Taken together, these results confirmed that GABAergic interneuron and synapse transmission are disturbed profoundly in an mTOR-dependent manner in type II FCD. Our study suggests that postsynaptic mechanisms independent of interneuron reduction or altered expression of GABA synapse genes might be accountable for the impaired GABAergic neurotransmission in type II FCD as well as other mTOR-related epilepsies.
Collapse
|
30
|
Boll I, Jensen P, Schwämmle V, Larsen MR. Depolarization-dependent Induction of Site-specific Changes in Sialylation on N-linked Glycoproteins in Rat Nerve Terminals. Mol Cell Proteomics 2020; 19:1418-1435. [PMID: 32518069 PMCID: PMC8143646 DOI: 10.1074/mcp.ra119.001896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/08/2020] [Indexed: 12/11/2022] Open
Abstract
Synaptic transmission leading to release of neurotransmitters in the nervous system is a fast and highly dynamic process. Previously, protein interaction and phosphorylation have been thought to be the main regulators of synaptic transmission. Here we show that sialylation of N-linked glycosylation is a novel potential modulator of neurotransmitter release mechanisms by investigating depolarization-dependent changes of formerly sialylated N-linked glycopeptides. We suggest that negatively charged sialic acids can be modulated, similarly to phosphorylation, by the action of sialyltransferases and sialidases thereby changing local structure and function of membrane glycoproteins. We characterized site-specific alteration in sialylation on N-linked glycoproteins in isolated rat nerve terminals after brief depolarization using quantitative sialiomics. We identified 1965 formerly sialylated N-linked glycosites in synaptic proteins and found that the abundances of 430 glycosites changed after 5 s depolarization. We observed changes on essential synaptic proteins such as synaptic vesicle proteins, ion channels and transporters, neurotransmitter receptors and cell adhesion molecules. This study is to our knowledge the first to describe ultra-fast site-specific modulation of the sialiome after brief stimulation of a biological system.
Collapse
Affiliation(s)
- Inga Boll
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Veit Schwämmle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
31
|
Han P, Welsh CT, Smith MT, Schmidt RE, Carroll SL. Complex Patterns of GABAergic Neuronal Deficiency and Type 2 Potassium-Chloride Cotransporter Immaturity in Human Focal Cortical Dysplasia. J Neuropathol Exp Neurol 2020; 78:365-372. [PMID: 30856249 DOI: 10.1093/jnen/nlz009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Focal cortical dysplasia (FCD) is a common histopathologic finding in cortical specimens resected for refractory epilepsy. GABAergic neuronal abnormalities and K-Cl cotransporter type 2 (KCC2) immaturity may be contributing factors for FCD-related epilepsy. We examined surgical specimens from 12 cases diagnosed with FCD, and brain tissues without developmental abnormality obtained from 6 autopsy cases. We found that GABAergic neuronal density was abnormal in FCD with 2 distinct patterns. In 7 of 12 (58%) FCD subjects, the GABAergic neuron density in dysplastic regions and in neighboring nondysplastic regions was equally reduced, hence we call this a "broad pattern." In the remaining cases, GABAergic neuron density was decreased in dysplastic regions but not in the neighboring nondysplastic regions; we designate this "restricted pattern." The different patterns are not associated with pathologic subtypes of FCD. Intracytoplasmic retention of KCC2 is evident in dysmorphic neurons in the majority of FCD type II subjects (5/7) but not in FCD type I. Our study suggests that (1) "broad" GABAergic deficiency may reflect epileptic vulnerability outside the dysplastic area; and (2) abnormal distribution of KCC2 may contribute to seizure generation in patients with FCD type II but not in type I.
Collapse
Affiliation(s)
- Pengcheng Han
- Department of Pathology and Laboratory Medicine.,Department of Pathology and Laboratory Medicine Residency Program, Medical University of South Carolina, Charleston, South Carolina
| | | | | | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | | |
Collapse
|
32
|
Subramanian L, Calcagnotto ME, Paredes MF. Cortical Malformations: Lessons in Human Brain Development. Front Cell Neurosci 2020; 13:576. [PMID: 32038172 PMCID: PMC6993122 DOI: 10.3389/fncel.2019.00576] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Creating a functional cerebral cortex requires a series of complex and well-coordinated developmental steps. These steps have evolved across species with the emergence of cortical gyrification and coincided with more complex behaviors. The presence of diverse progenitor cells, a protracted timeline for neuronal migration and maturation, and diverse neuronal types are developmental features that have emerged in the gyrated cortex. These factors could explain how the human brain has expanded in size and complexity. However, their complex nature also renders new avenues of vulnerability by providing additional cell types that could contribute to disease and longer time windows that could impact the composition and organization of the cortical circuit. We aim to discuss the unique developmental steps observed in human corticogenesis and propose how disruption of these species-unique processes could lead to malformations of cortical development.
Collapse
Affiliation(s)
- Lakshmi Subramanian
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mercedes F Paredes
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States.,Neuroscience Graduate Division, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
33
|
Represa A. Why Malformations of Cortical Development Cause Epilepsy. Front Neurosci 2019; 13:250. [PMID: 30983952 PMCID: PMC6450262 DOI: 10.3389/fnins.2019.00250] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/04/2019] [Indexed: 12/13/2022] Open
Abstract
Malformations of cortical development (MCDs), a complex family of rare disorders, result from alterations of one or combined developmental steps, including progenitors proliferation, neuronal migration and differentiation. They are an important cause of childhood epilepsy and frequently associate cognitive deficits and behavioral alterations. Though the physiopathological mechanisms of epilepsy in MCD patients remain poorly elucidated, research during the past decade highlighted the contribution of some factors that will be reviewed in this paper and that include: (i) the genes that caused the malformation, that can be responsible for a significant reduction of inhibitory cells (e.g., ARX gene) or be inducing cell-autonomous epileptogenic changes in affected neurons (e.g., mutations on the mTOR pathway); (ii) the alteration of cortical networks development induced by the malformation that will also involve adjacent or distal cortical areas apparently sane so that the epileptogenic focus might be more extended that the malformation or even localized at distance from it; (iii) the normal developmental processes that would influence and determine the onset of epilepsy in MCD patients, particularly precocious in most of the cases.
Collapse
Affiliation(s)
- Alfonso Represa
- INSERM, Institut de Neurobiologie de la Méditerranée, Aix-Marseille University, Marseille, France
| |
Collapse
|
34
|
Blauwblomme T, Dossi E, Pellegrino C, Goubert E, Iglesias BG, Sainte-Rose C, Rouach N, Nabbout R, Huberfeld G. Gamma-aminobutyric acidergic transmission underlies interictal epileptogenicity in pediatric focal cortical dysplasia. Ann Neurol 2019; 85:204-217. [PMID: 30597612 DOI: 10.1002/ana.25403] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Dysregulation of γ-aminobutyric acidergic (GABAergic) transmission has been reported in lesional acquired epilepsies (gliomas, hippocampal sclerosis). We investigated its involvement in a developmental disorder, human focal cortical dysplasia (FCD), focusing on chloride regulation driving GABAergic signals. METHODS In vitro recordings of 47 human cortical acute slices from 11 pediatric patients who received operations for FCD were performed on multielectrode arrays. GABAergic receptors and chloride regulators were pharmacologically modulated. Immunostaining for chloride cotransporter KCC2 and interneurons were performed on recorded slices to correlate electrophysiology and expression patterns. RESULTS FCD slices retain intrinsic epileptogenicity. Thirty-six of 47 slices displayed spontaneous interictal discharges, along with a pattern specific to the histological subtypes. Ictal discharges were induced in proepileptic conditions in 6 of 8 slices in the areas generating spontaneous interictal discharges, with a transition to seizure involving the emergence of preictal discharges. Interictal discharges were sustained by GABAergic signaling, as a GABAA receptor blocker stopped them in 2 of 3 slices. Blockade of NKCC1 Cl- cotransporters further controlled interictal discharges in 9 of 12 cases, revealing a Cl- dysregulation affecting actions of GABA. Immunohistochemistry highlighted decreased expression and changes in KCC2 subcellular localization and a decrease in the number of GAD67-positive interneurons in regions generating interictal discharges. INTERPRETATION Altered chloride cotransporter expression and changes in interneuron density in FCD may lead to paradoxical depolarization of pyramidal cells. Spontaneous interictal discharges are consequently mediated by GABAergic signals, and targeting chloride regulation in neurons may be considered for the development of new antiepileptic drugs. Ann Neurol 2019; 1-14 ANN NEUROL 2019;85:204-217.
Collapse
Affiliation(s)
- Thomas Blauwblomme
- APHP, Department of Pediatric Neurosurgery, Hospital Necker, Paris, France.,Université René Descartes. PRES Sorbonne Paris Cité, Paris, France.,INSERM U1129, Infantile Epilepsies and Brain Plasticity, Paris Descartes University, PRES Sorbonne Paris Cité, Paris, France
| | - Elena Dossi
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | | | | | - Beatriz Gal Iglesias
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea, Madrid, Spain
| | - Christian Sainte-Rose
- APHP, Department of Pediatric Neurosurgery, Hospital Necker, Paris, France.,Université René Descartes. PRES Sorbonne Paris Cité, Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Rima Nabbout
- Université René Descartes. PRES Sorbonne Paris Cité, Paris, France.,APHP, Department of Neuropediatrics, Hospital Necker, Paris, France
| | - Gilles Huberfeld
- INSERM U1129, Infantile Epilepsies and Brain Plasticity, Paris Descartes University, PRES Sorbonne Paris Cité, Paris, France.,Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNR UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.,Sorbonne University, AP-HP, Department of Neurophysiology, La Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
35
|
Martinez-Lizana E, Fauser S, Brandt A, Schuler E, Wiegand G, Doostkam S, San Antonio-Arce V, Jacobs J, Bast T, Shah M, Zentner J, Schulze-Bonhage A. Long-term seizure outcome in pediatric patients with focal cortical dysplasia undergoing tailored and standard surgical resections. Seizure 2018; 62:66-73. [PMID: 30296740 DOI: 10.1016/j.seizure.2018.09.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Focal cortical dysplasia (FCD) is the major cause of focal intractable epilepsy in childhood. Here we analyze the factors influencing the success of surgical treatment in a large cohort of children with histologically ascertained FCD. METHOD A retrospective study of the effects of FCD type, surgical intervention, and age at surgery in a pediatric cohort. RESULTS A total of 113 patients (71 male; mean age at surgery 10.3 years; range 0-18) were analyzed; 45 had undergone lesionectomy, 42 lobectomy, 18 multi-lobectomy, and eight hemispherotomy. Complete seizure control (Engel Ia) was achieved in 56% after two years, 52% at five years, and 50% at last follow-up (18-204 months). Resections were more extensive in younger patients (40% of the surgeries affecting more than one lobe in patients aged nine years or younger vs. 22% in patients older than nine years). While resections were more limited in older children, their long-term outcome tended to be superior (42% seizure freedom in patients aged nine years or younger vs. 56% in patients older than nine years). The outcome in FCD I was not significantly inferior to that in FCD II. CONCLUSIONS Our data confirm the long-term efficacy of surgery in children with FCD and epilepsy. An earlier age at surgery within this cohort did not predict a better long-term outcome, but it involved less-tailored surgical approaches. The data suggest that in patients with an unclear extent of the dysplastic area, later resections may offer advantages in terms of the precision of surgical-resection planning.
Collapse
Affiliation(s)
- Eva Martinez-Lizana
- Dept. of Epileptology, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany.
| | | | - Armin Brandt
- Dept. of Epileptology, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | | | - Gert Wiegand
- Dept. of Pediatric Neurology, University of Kiel, Kiel, Germany
| | - Soroush Doostkam
- Faculty of Medicine, University of Freiburg, Germany; Institute of Neuropathology, Medical Center - University of Freiburg, Germany
| | - Victoria San Antonio-Arce
- Dept. of Epileptology, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; Dept. of Pediatric Neurology, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Julia Jacobs
- Dept. of Epileptology, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - Thomas Bast
- Faculty of Medicine, University of Freiburg, Germany; Epilepsy Center Kork, Germany
| | - Mukesch Shah
- Faculty of Medicine, University of Freiburg, Germany; Dept. Neurosurgery, Medical Center - University of Freiburg, Germany
| | - Josef Zentner
- Faculty of Medicine, University of Freiburg, Germany; Dept. Neurosurgery, Medical Center - University of Freiburg, Germany
| | - Andreas Schulze-Bonhage
- Dept. of Epileptology, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
36
|
Challenges in managing epilepsy associated with focal cortical dysplasia in children. Epilepsy Res 2018; 145:1-17. [DOI: 10.1016/j.eplepsyres.2018.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 04/30/2018] [Accepted: 05/12/2018] [Indexed: 12/15/2022]
|
37
|
Fujimoto A, Okanishi T, Kanai S, Sato K, Itamura S, Baba S, Nishimura M, Masui T, Enoki H. Double match of 18F-fluorodeoxyglucose-PET and iomazenil-SPECT improves outcomes of focus resection surgery. Acta Neurochir (Wien) 2018; 160:1875-1882. [PMID: 29858947 DOI: 10.1007/s00701-018-3573-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 05/23/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND When the results of electroencephalography (EEG), magnetic resonance imaging (MRI), and seizure semiology are discordant or no structural lesion is evident on MRI, single-photon emission computed tomography (SPECT) and positron emission tomography (PET) are important examinations for lateralization or localization of epileptic regions. We hypothesized that the concordance between interictal 2-[18F]fluoro-2-deoxy-D-glucose (18FDG)-PET and iomazenil (IMZ)-SPECT could suggest the epileptogenic lobe in patients with non-lesional findings on MRI. METHOD Fifty-nine patients (31 females, 28 males; mean age, 29 years; median age, 27 years; range, 7-56 years) underwent subdural electrode implantation followed by focus resection. All patients underwent 18FDG-PET, IMZ-SPECT, and focus resection surgery. Follow-up was continued for ≥ 2 years. We evaluated surgical outcomes as seizure-free or not and analyzed correlations between outcomes and concordances of low-uptake lobes on PET, SPECT, or both PET and SPECT to the resection lobes. We used uni- and multivariate logistic regression analyses. RESULTS In univariate analyses, all three concordances correlated significantly with seizure-free outcomes (PET, p = 0.017; SPECT, p = 0.030; both PET and SPECT, p = 0.006). In multivariate analysis, concordance between resection and low-uptake lobes in both PET and SPECT correlated significantly with seizure-free outcomes (p = 0.004). The odds ratio was 6.0. CONCLUSION Concordance between interictal 18FDG-PET and IMZ-SPECT suggested that the epileptogenic lobe is six times better than each examination alone among patients with non-lesional findings on MRI. IMZ-SPECT and 18FDG-PET are complementary examinations in the assessment of localization-related epilepsy.
Collapse
|
38
|
Hu S, Knowlton RC, Watson BO, Glanowska KM, Murphy GG, Parent JM, Wang Y. Somatic Depdc5 deletion recapitulates electroclinical features of human focal cortical dysplasia type IIA. Ann Neurol 2018; 84:140-146. [PMID: 30080265 PMCID: PMC6119494 DOI: 10.1002/ana.25272] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/02/2018] [Accepted: 06/10/2018] [Indexed: 01/04/2023]
Abstract
Epileptogenic mechanisms in focal cortical dysplasia (FCD) remain elusive, as no animal models faithfully recapitulate FCD seizures, which have distinct electrographic features and a wide range of semiologies. Given that DEPDC5 plays significant roles in focal epilepsies with FCD, we used in utero electroporation with clustered regularly interspaced short palindromic repeats gene deletion to create focal somatic Depdc5 deletion in the rat embryonic brain. Animals developed spontaneous seizures with focal pathological and electroclinical features highly clinically relevant to FCD IIA, paving the way toward understanding its pathogenesis and developing mechanistic-based therapies. Ann Neurol 2018;83:140-146.
Collapse
Affiliation(s)
- Shuntong Hu
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Robert C. Knowlton
- Department of Neurology, University of California San Francisco, San Francisco, CA
| | | | - Katarzyna M. Glanowska
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI
| | - Geoffrey G. Murphy
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI
| | - Jack M. Parent
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Ann Arbor VA Healthcare System, Ann Arbor, MI
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
39
|
Deshmukh A, Leichner J, Bae J, Song Y, Valdés-Hernández PA, Lin WC, Riera JJ. Histological Characterization of the Irritative Zones in Focal Cortical Dysplasia Using a Preclinical Rat Model. Front Cell Neurosci 2018; 12:52. [PMID: 29867355 PMCID: PMC5968101 DOI: 10.3389/fncel.2018.00052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/15/2018] [Indexed: 12/19/2022] Open
Abstract
Current clinical practice in focal epilepsy involves brain source imaging (BSI) to localize brain areas where from interictal epileptiform discharges (IEDs) emerge. These areas, named irritative zones, have been useful to define candidate seizures-onset zones during pre-surgical workup. Since human histological data are mostly available from final resected zones, systematic studies characterizing pathophysiological mechanisms and abnormal molecular/cellular substrates in irritative zones—independent of them being epileptogenic—are challenging. Combining BSI and histological analysis from all types of irritative zones is only possible through the use of preclinical animal models. Here, we recorded 32-channel spontaneous electroencephalographic data from rats that have focal cortical dysplasia (FCD) and chronic seizures. BSI for different IED subtypes was performed using the methodology presented in Bae et al. (2015). Post-mortem brain sections containing irritative zones were stained to quantify anatomical, functional, and inflammatory biomarkers specific for epileptogenesis, and the results were compared with those obtained using the contralateral healthy brain tissue. We found abnormal anatomical structures in all irritative zones (i.e., larger neuronal processes, glioreactivity, and vascular cuffing) and larger expressions for neurotransmission (i.e., NR2B) and inflammation (i.e., ILβ1, TNFα and HMGB1). We conclude that irritative zones in this rat preclinical model of FCD comprise abnormal tissues disregarding whether they are actually involved in icto-genesis or not. We hypothesize that seizure perpetuation happens gradually; hence, our results could support the use of IED-based BSI for the early diagnosis and preventive treatment of potential epileptic foci. Further verifications in humans are yet needed.
Collapse
Affiliation(s)
- Abhay Deshmukh
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jared Leichner
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jihye Bae
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Yinchen Song
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Pedro A Valdés-Hernández
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Wei-Chiang Lin
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jorge J Riera
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| |
Collapse
|
40
|
Kielbinski M, Setkowicz Z, Gzielo K, Janeczko K. Profiles of gene expression in the hippocampal formation of rats with experimentally-induced brain dysplasia. Dev Neurobiol 2018; 78:718-735. [DOI: 10.1002/dneu.22595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/28/2018] [Accepted: 04/06/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Michal Kielbinski
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Kinga Gzielo
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| | - Krzysztof Janeczko
- Department of Neuroanatomy; Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9; Krakow 30-387 Poland
| |
Collapse
|
41
|
Bartoli A, Tyrand R, Vargas MI, Momjian S, Boëx C. Low Frequency Microstimulation Is Locally Excitatory in Patients With Epilepsy. Front Neural Circuits 2018; 12:22. [PMID: 29670511 PMCID: PMC5893788 DOI: 10.3389/fncir.2018.00022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
Deep brain stimulation (DBS) could become a palliative treatment for patients with drug-resistant epilepsy for which surgery cannot be proposed. The objective of this study was to perform microstimulation to measure the effects of DBS in epilepsy locally at the level of a few neurons, with microelectrode recordings, for the first time in patients with epilepsy. Microelectrode recordings were performed before, during and after microstimulation in nine patients with refractory epilepsy. Neuronal spikes were successfully extracted from multi-unit recordings with clustering in six out of seven patients during hippocampal and in one out of two patients during cortical dysplasia microstimulation (1 Hz, charge-balanced biphasic waveform, 60 μs/ph, 25 μA). The firing rates increased in four out of the six periods of microstimulation that could be analyzed. The firing rates were found higher than before microstimulation in all eight periods with increases reaching significance in six out of eight periods. Low-frequency microstimulation was hence sufficient to induce neuronal excitation lasting beyond the stimulation period. No inhibition was observed. This report presents the first evidence that microstimulation performed in epileptic patients produced locally neuronal excitation. Hence neuronal excitation is shown here as the local mechanism of action of DBS. This local excitation is in agreement with epileptogenic effects of low-frequency hippocampal macrostimulation.
Collapse
Affiliation(s)
- Andrea Bartoli
- Department of Neurosurgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Rémi Tyrand
- Department of Neurology, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maria I Vargas
- Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Neuroradiology, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Shahan Momjian
- Department of Neurosurgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Colette Boëx
- Department of Neurology, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
42
|
Kielbinski M, Gzielo K, Soltys Z. Review: Roles for astrocytes in epilepsy: insights from malformations of cortical development. Neuropathol Appl Neurobiol 2018; 42:593-606. [PMID: 27257021 DOI: 10.1111/nan.12331] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/25/2016] [Accepted: 06/03/2016] [Indexed: 12/25/2022]
Abstract
Malformations of cortical development (MCDs), such as cortical dysplasia and tuberous sclerosis complex, are common causes of intractable epilepsy, especially in paediatric patients. Recently, mounting evidence points to a common pathology of these disorders. Hyperactivation of mammalian target of rapamycin (mTOR) has been proposed as a central mechanism in most, if not all, MCDs. The transition from mTOR hyperactivation and cellular abnormalities to large-scale functional changes and seizure is, however, not fully understood. In this article we set out to review currently available information regarding MCD pathology, focusing on glial cells - especially astrocytes - and their interactions with the brain vascular system. A large body of evidence points to these elements as potential targets in MCD. Here, we attempt to provide a review of this evidence and propose some hypotheses regarding the possible chain of events linking primary glial dysfunction and epilepsy. We focus on extracellular matrix remodelling, blood-brain barrier leakage and failure of astrocyte-dependent removal of extracellular debris. We posit that the failure of these systems results in a chronically pro-inflammatory environment, maintaining local astrocytes in a state of gliosis, with increased susceptibility to seizures as a consequence.
Collapse
Affiliation(s)
- M Kielbinski
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - K Gzielo
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Z Soltys
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
43
|
Hernan AE, Mahoney JM, Curry W, Richard G, Lucas MM, Massey A, Holmes GL, Scott RC. Environmental enrichment normalizes hippocampal timing coding in a malformed hippocampus. PLoS One 2018; 13:e0191488. [PMID: 29394267 PMCID: PMC5796690 DOI: 10.1371/journal.pone.0191488] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/05/2018] [Indexed: 12/24/2022] Open
Abstract
Neurodevelopmental insults leading to malformations of cortical development (MCD) are a common cause of psychiatric disorders, learning impairments and epilepsy. In the methylazoxymethanol (MAM) model of MCDs, animals have impairments in spatial cognition that, remarkably, are improved by post-weaning environmental enrichment (EE). To establish how EE impacts network-level mechanisms of spatial cognition, hippocampal in vivo single unit recordings were performed in freely moving animals in an open arena. We took a generalized linear modeling approach to extract fine spike timing (FST) characteristics and related these to place cell fidelity used as a surrogate of spatial cognition. We find that MAM disrupts FST and place-modulated rate coding in hippocampal CA1 and that EE improves many FST parameters towards normal. Moreover, FST parameters predict spatial coherence of neurons, suggesting that mechanisms determining altered FST are responsible for impaired cognition in MCDs. This suggests that FST parameters could represent a therapeutic target to improve cognition even in the context of a brain that develops with a structural abnormality.
Collapse
Affiliation(s)
- Amanda E. Hernan
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
- * E-mail: (RCS); (AEH)
| | - J. Matthew Mahoney
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Willie Curry
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Greg Richard
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Marcella M. Lucas
- Department of Neurology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Andrew Massey
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Gregory L. Holmes
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Rod C. Scott
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, United States of America
- University College London, Institute of Child Health, London, United Kingdom
- * E-mail: (RCS); (AEH)
| |
Collapse
|
44
|
Iffland PH, Crino PB. Focal Cortical Dysplasia: Gene Mutations, Cell Signaling, and Therapeutic Implications. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:547-571. [PMID: 28135561 DOI: 10.1146/annurev-pathol-052016-100138] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Focal cortical dysplasias (FCDs) are malformations of cortical development (MCDs) that are highly associated with medication-resistant epilepsy and are the most common cause of neocortical epilepsy in children. FCDs are a heterogeneous group of developmental disorders caused by germline or somatic mutations that occur in genes regulating the PI3K/Akt/mTOR pathway-a key pathway in neuronal growth and migration. Accordingly, FCDs are characterized by abnormal cortical lamination, cell morphology (e.g., cytomegaly), and cellular polarity. In some FCD subtypes, balloon cells express proteins typically seen in neuroglial progenitor cells. Because recurrent intractable seizures are a common feature of FCDs, epileptogenic electrophysiological properties are also observed in addition to local inflammation. Here, we will summarize the current literature regarding FCDs, addressing the current classification system, histopathology, molecular genetics, electrophysiology, and transcriptome and cell signaling changes.
Collapse
Affiliation(s)
- Philip H Iffland
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140;
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland 21201;
| |
Collapse
|
45
|
Vendramin Pasquetti M, Meier L, Loureiro S, Ganzella M, Junges B, Barbieri Caus L, Umpierrez Amaral A, Koeller DM, Goodman S, Woontner M, Gomes de Souza DO, Wajner M, Calcagnotto ME. Impairment of GABAergic system contributes to epileptogenesis in glutaric acidemia type I. Epilepsia 2017; 58:1771-1781. [PMID: 28762469 DOI: 10.1111/epi.13862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Glutaric acidemia type I (GA-I) is an inherited neurometabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH) and characterized by increased levels of glutaric, 3-OH-glutaric, and glutaconic acids in the brain parenchyma. The increment of these organic acids inhibits glutamate decarboxylase (GAD) and consequently lowers the γ-aminobutyric acid (GABA) synthesis. Untreated patients exhibit severe neurologic deficits during development, including epilepsy, especially following an acute encephalopathy outbreak. In this work, we evaluated the role of the GABAergic system on epileptogenesis in GA-I using the Gcdh-/- mice exposed to a high lysine diet (Gcdh-/- -Lys). METHODS Spontaneous recurrent seizures (SRS), seizure susceptibility, and changes in brain oscillations were evaluated by video-electroencephalography (EEG). Cortical GABAergic synaptic transmission was evaluated using electrophysiologic and neurochemical approaches. RESULTS SRS were observed in 72% of Gcdh-/- -Lys mice, whereas no seizures were detected in age-matched controls (Gcdh+/+ or Gcdh-/- receiving normal diet). The severity and number of PTZ-induced seizures were higher in Gcdh-/- -Lys mice. EEG spectral analysis showed a significant decrease in theta and gamma oscillations and predominant delta waves in Gcdh-/- -Lys mice, associated with increased EEG left index. Analysis of cortical synaptosomes revealed a significantly increased percentage of glutamate release and decreased GABA release in Gcdh-/- -Lys mice that were associated with a decrease in cortical GAD immunocontent and activity and confirmed by reduced frequency of inhibitory events in cortical pyramidal cells. SIGNIFICANCE Using an experimental model with a phenotype similar to that of GA-I in humans-the Gcdh-/- mice under high lysine diet (Gcdh-/- -Lys)-we provide evidence that a reduction in cortical inhibition of Gcdh-/- -Lys mice, probably induced by GAD dysfunction, leads to hyperexcitability and increased slow oscillations associated with neurologic abnormalities in GA-I. Our findings offer a new perspective on the pathophysiology of brain damage in GA-I.
Collapse
Affiliation(s)
- Mayara Vendramin Pasquetti
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory-NNNESP Lab.), Biochemistry Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Letícia Meier
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory-NNNESP Lab.), Biochemistry Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Samanta Loureiro
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcelo Ganzella
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bernardo Junges
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory-NNNESP Lab.), Biochemistry Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Letícia Barbieri Caus
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory-NNNESP Lab.), Biochemistry Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre Umpierrez Amaral
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - David M Koeller
- Department of Pediatrics, Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, U.S.A
| | - Stephen Goodman
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, U.S.A
| | - Michael Woontner
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, U.S.A
| | - Diogo Onofre Gomes de Souza
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Moacir Wajner
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Elisa Calcagnotto
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory-NNNESP Lab.), Biochemistry Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Postgraduate Program in Neuroscience, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
46
|
Medial Ganglionic Eminence Progenitors Transplanted into Hippocampus Integrate in a Functional and Subtype-Appropriate Manner. eNeuro 2017; 4:eN-NWR-0359-16. [PMID: 28413826 PMCID: PMC5388838 DOI: 10.1523/eneuro.0359-16.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Medial ganglionic eminence (MGE) transplantation rescues disease phenotypes in various preclinical models with interneuron deficiency or dysfunction, including epilepsy. While underlying mechanism(s) remains unclear to date, a simple explanation is that appropriate synaptic integration of MGE-derived interneurons elevates GABA-mediated inhibition and modifies the firing activity of excitatory neurons in the host brain. However, given the complexity of interneurons and potential for transplant-derived interneurons to integrate or alter the host network in unexpected ways, it remains unexplored whether synaptic connections formed by transplant-derived interneurons safely mirror those associated with endogenous interneurons. Here, we combined optogenetics, interneuron-specific Cre driver mouse lines, and electrophysiology to study synaptic integration of MGE progenitors. We demonstrated that MGE-derived interneurons, when transplanted into the hippocampus of neonatal mice, migrate in the host brain, differentiate to mature inhibitory interneurons, and form appropriate synaptic connections with native pyramidal neurons. Endogenous and transplant-derived MGE progenitors preferentially formed inhibitory synaptic connections onto pyramidal neurons but not endogenous interneurons. These findings demonstrate that transplanted MGE progenitors functionally integrate into the postnatal hippocampal network.
Collapse
|
47
|
Glial GABA Transporters as Modulators of Inhibitory Signalling in Epilepsy and Stroke. ADVANCES IN NEUROBIOLOGY 2017; 16:137-167. [PMID: 28828609 DOI: 10.1007/978-3-319-55769-4_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Imbalances in GABA-mediated tonic inhibition are involved in several pathophysiological conditions. A classical way of controlling tonic inhibition is through pharmacological intervention with extrasynaptic GABAA receptors that sense ambient GABA and mediate a persistent GABAergic conductance. An increase in tonic inhibition may, however, also be obtained indirectly by inhibiting glial GABA transporters (GATs). These are sodium-coupled membrane transport proteins that normally act to terminate GABA neurotransmitter action by taking up GABA into surrounding astrocytes. The aim of the review is to provide an overview of glial GATs in regulating tonic inhibition, especially in epilepsy and stroke. This entails a comprehensive summary of changes known to occur in GAT expression levels and signalling following epileptic and ischemic insults. Further, we discuss the accumulating pharmacological evidence for targeting GATs in these diseases.
Collapse
|
48
|
Siedlecka M, Grajkowska W, Galus R, Dembowska-Bagińska B, Jóźwiak J. Focal cortical dysplasia: Molecular disturbances and clinicopathological classification (Review). Int J Mol Med 2016; 38:1327-1337. [DOI: 10.3892/ijmm.2016.2760] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/23/2016] [Indexed: 11/05/2022] Open
|
49
|
Zakharova I, Kohlmeyer JC, Kornhuber ME. Facilitation dynamics of late somatosensory evoked potentials after sural nerve stimulation. Clin Neurophysiol 2016; 127:2545-50. [PMID: 27291872 DOI: 10.1016/j.clinph.2016.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 03/06/2016] [Accepted: 04/04/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Somatosensory evoked potentials (SSEPs) could be suitable for elucidating the properties of synaptic potentials (SPs). Two experiments were designed for this purpose. METHODS 1st experiment: the sural nerve was stimulated in 13 subjects with single or trains of 3 stimuli (1Hz or 0.4Hz), the within train interstimulus interval (ISI) was stepwise extended from 2 to 10ms. Cz' against Fz, time interval 500ms. 2nd experiment: Gating was investigated in a paired stimulus paradigm with intervals of 0.7, 1, 2, 5s in 15 subjects after single and train stimuli (ISI 3ms) with equal stimulus and recording positions. RESULTS 1st experiment: N1-P1, P1-N2a, and P2-N2b but not N37-P40 displayed a significant gain in amplitude following train stimuli compared with single stimuli. Significantly larger N1-P1 amplitude values were observed with 0.4Hz stimulus repetition compared with 1.0Hz. Short ISIs of 2-4ms led to higher N1-P1 amplitudes than obtained with longer ISIs of 7-10ms. 2nd experiment: recovery of the habituated N1-P1 amplitude was complete when the 2nd of 2 stimuli followed after 2s. CONCLUSIONS SSEP vertex potential amplitudes (especially N1-P1) recorded after train stimuli presumably reflect the decay dynamics of excitatory postsynaptic potentials. Recovery of the habituated N1 (2nd experiment) was complete within 2s. SIGNIFICANCE Our study may be relevant to study properties of excitatory synaptic potentials in diseases of the central nervous system such as e.g. epilepsy or migraine.
Collapse
Affiliation(s)
- I Zakharova
- Dept. of Neurology, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - J C Kohlmeyer
- Dept. of Neurology, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - M E Kornhuber
- Dept. of Neurology, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany.
| |
Collapse
|
50
|
Medici V, Rossini L, Deleo F, Tringali G, Tassi L, Cardinale F, Bramerio M, de Curtis M, Garbelli R, Spreafico R. Different parvalbumin and GABA expression in human epileptogenic focal cortical dysplasia. Epilepsia 2016; 57:1109-19. [DOI: 10.1111/epi.13405] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Valentina Medici
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| | - Laura Rossini
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| | - Francesco Deleo
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| | - Giovanni Tringali
- Department of Neurosurgery, IRCCS; Foundation Neurological Institute “C. Besta”; Milan Italy
| | - Laura Tassi
- Epilepsy Surgery Center C. Munari; Milan Italy
| | | | | | - Marco de Curtis
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| | - Rita Garbelli
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| | - Roberto Spreafico
- Clinical Epileptology and Experimental Neurophysiology Unit; Istituto Neurologico “C. Besta”; Milan Italy
| |
Collapse
|