1
|
Kazis D, Chatzikonstantinou S, Ciobica A, Kamal FZ, Burlui V, Calin G, Mavroudis I. Epidemiology, Risk Factors, and Biomarkers of Post-Traumatic Epilepsy: A Comprehensive Overview. Biomedicines 2024; 12:410. [PMID: 38398011 PMCID: PMC10886732 DOI: 10.3390/biomedicines12020410] [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: 01/23/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
This paper presents an in-depth exploration of Post-Traumatic Epilepsy (PTE), a complex neurological disorder following traumatic brain injury (TBI), characterized by recurrent, unprovoked seizures. With TBI being a global health concern, understanding PTE is crucial for effective diagnosis, management, and prognosis. This study aims to provide a comprehensive overview of the epidemiology, risk factors, and emerging biomarkers of PTE, thereby informing clinical practice and guiding future research. The epidemiological aspect of the study reveals PTE as a significant contributor to acquired epilepsies, with varying incidence influenced by injury severity, age, and intracranial pathologies. The paper delves into the multifactorial nature of PTE risk factors, encompassing clinical, demographic, and genetic elements. Key insights include the association of injury severity, intracranial hemorrhages, and early seizures with increased PTE risk, and the roles of age, gender, and genetic predispositions. Advancements in neuroimaging, electroencephalography, and molecular biology are presented, highlighting their roles in identifying potential PTE biomarkers. These biomarkers, ranging from radiological signs to electroencephalography EEG patterns and molecular indicators, hold promise for enhancing PTE pathogenesis understanding, early diagnosis, and therapeutic guidance. The paper also discusses the critical roles of astrocytes and microglia in PTE, emphasizing the significance of neuroinflammation in PTE development. The insights from this review suggest potential therapeutic targets in neuroinflammation pathways. In conclusion, this paper synthesizes current knowledge in the field, emphasizing the need for continued research and a multidisciplinary approach to effectively manage PTE. Future research directions include longitudinal studies for a better understanding of TBI and PTE outcomes, and the development of targeted interventions based on individualized risk profiles. This research contributes significantly to the broader understanding of epilepsy and TBI.
Collapse
Affiliation(s)
- Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 20th Carol I Avenue, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
| | - Fatima Zahra Kamal
- Higher Institute of Nursing Professions and Health Technical (ISPITS), Marrakech 40000, Morocco
- Laboratory of Physical Chemistry of Processes and Materials, Faculty of Sciences and Techniques, Hassan First University, Settat 26000, Morocco
| | - Vasile Burlui
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Gabriela Calin
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, Leeds LS2 9JT, UK
- Faculty of Medicine, Leeds University, Leeds LS2 9JT, UK
| |
Collapse
|
2
|
Yang X, Li T, Liu J, Sun H, Cheng L, Song X, Han Z, Luo H, Han W, Xie L, Jiang L. Effects of minocycline on dendrites, dendritic spines, and microglia in immature mouse brains after kainic acid-induced status epilepticus. CNS Neurosci Ther 2024; 30:e14352. [PMID: 37438982 PMCID: PMC10848062 DOI: 10.1111/cns.14352] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023] Open
Abstract
PURPOSE This study aimed to investigate whether minocycline could influence alterations of microglial subtypes, the morphology of dendrites and dendritic spines, the microstructures of synapses and synaptic proteins, or even cognition outcomes in immature male mice following status epilepticus (SE) induced by kainic acid. METHODS Golgi staining was performed to visualize the dendrites and dendritic spines of neurons of the hippocampus. The microstructures of synapses and synaptic proteins were observed using transmission electron microscopy and western blotting analysis, respectively. Microglial reactivation and their markers were evaluated using flow cytometry. The Morris water maze (MWM) test was used to analyze spatial learning and memory ability. RESULTS Significant partial spines increase (predominate in thin spines) was observed in the dendrites of neurons after acute SE and partial loss (mainly in thin spines) was presented by days 14 and 28 post-SE. The postsynaptic ultrastructure was impaired on the 7th and 14th days after SE. The proportion of M1 microglia increased significantly only after acute SE Similarly, the proportion of M2 microglia increased in the acute stage with high expression levels of all surface markers. In contrast, a decrease in M2 microglia and their markers was noted by day 14 post-SE. Minocycline could reverse the changes in dendrites and synaptic proteins caused by SE, and increase the levels of synaptic proteins. Meanwhile, minocycline could inhibit the reactivation of M1 microglia and the expression of their markers, except for promoting CD200R. In addition, treatment with minocycline could regulate the expression of M2 microglia and their surface markers, as well as ameliorating the impaired spatial learning and memory on the 28th day after SE. CONCLUSIONS Dendritic spines and microglia are dynamically changed after SE. Minocycline could ameliorate the impaired cognition in the kainic acid-induced mouse model by decreasing the damage to dendrites and altering microglial reactivation.
Collapse
Affiliation(s)
- Xiaoyue Yang
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Tianyi Li
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Jie Liu
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Hong Sun
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Li Cheng
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Xiaojie Song
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Ziyao Han
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Hanyu Luo
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Wei Han
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Lingling Xie
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| | - Li Jiang
- Department of NeurologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
- National Clinical Research Center for Child Health and DisordersChongqingChina
- Ministry of Education Key Laboratory of Child Development and DisordersChongqingChina
- Chongqing Key Laboratory of PediatricsChongqingChina
| |
Collapse
|
3
|
Latchney SE, Ruiz Lopez BR, Womble PD, Blandin KJ, Lugo JN. Neuronal deletion of phosphatase and tensin homolog in mice results in spatial dysregulation of adult hippocampal neurogenesis. Front Mol Neurosci 2023; 16:1308066. [PMID: 38130682 PMCID: PMC10733516 DOI: 10.3389/fnmol.2023.1308066] [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: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Adult neurogenesis is a persistent phenomenon in mammals that occurs in select brain structures in both healthy and diseased brains. The tumor suppressor gene, phosphatase and tensin homolog deleted on chromosome 10 (Pten) has previously been found to restrict the proliferation of neural stem/progenitor cells (NSPCs) in vivo. In this study, we aimed to provide a comprehensive picture of how conditional deletion of Pten may regulate the genesis of adult NSPCs in the dentate gyrus of the hippocampus and the subventricular zone bordering the lateral ventricles. Using conventional markers and stereology, we quantified multiple stages of neurogenesis, including proliferating cells, immature neurons (neuroblasts), and apoptotic cells in several regions of the dentate gyrus, including the subgranular zone (SGZ), outer granule cell layer (oGCL), molecular layer, and hilus at 4 and 10 weeks of age. Our data demonstrate that conditional deletion of Pten in mice produces successive increases in dentate gyrus proliferating cells and immature neuroblasts, which confirms the known negative roles Pten has on cell proliferation and maturation. Specifically, we observe a significant increase in Ki67+ proliferating cells in the neurogenic SGZ at 4 weeks of age, but not 10 weeks of age. We also observe a delayed increase in neuroblasts at 10 weeks of age. However, our study expands on previous work by providing temporal, subregional, and neurogenesis-stage resolution. Specifically, we found that Pten deletion initially increases cell proliferation in the neurogenic SGZ, but this increase spreads to non-neurogenic dentate gyrus areas, including the hilus, oGCL, and molecular layer, as mice age. We also observed region-specific increases in apoptotic cells in the dentate gyrus hilar region that paralleled the regional increases in Ki67+ cells. Our work is accordant with the literature showing that Pten serves as a negative regulator of dentate gyrus neurogenesis but adds temporal and spatial components to the existing knowledge.
Collapse
Affiliation(s)
- Sarah E. Latchney
- Department of Biology, St. Mary’s College of Maryland, St. Mary’s City, MD, United States
| | - Brayan R. Ruiz Lopez
- Department of Biology, St. Mary’s College of Maryland, St. Mary’s City, MD, United States
| | - Paige D. Womble
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Katherine J. Blandin
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Joaquin N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| |
Collapse
|
4
|
Beesley S, Kumar SS. The t-N-methyl-d-aspartate receptor: Making the case for d-Serine to be considered its inverse co-agonist. Neuropharmacology 2023:109654. [PMID: 37437688 DOI: 10.1016/j.neuropharm.2023.109654] [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: 04/27/2023] [Revised: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
The N-methyl-d-aspartate receptor (NMDAR) is an enigmatic macromolecule that has garnered a good deal of attention on account of its involvement in the cellular processes that underlie learning and memory, following its discovery in the mid twentieth century (Baudry and Davis, 1991). Yet, despite advances in knowledge about its function, there remains much more to be uncovered regarding the receptor's biophysical properties, subunit composition, and role in CNS physiology and pathophysiology. The motivation for this review stems from the need for synthesizing new information gathered about these receptors that sheds light on their role in synaptic plasticity and their dichotomous relationship with the amino acid d-serine through which they influence the pathogenesis of neurodegenerative diseases like temporal lobe epilepsy (TLE), the most common type of adult epilepsies (Beesley et al., 2020a). This review will outline pertinent ideas relating structure and function of t-NMDARs (GluN3 subunit-containing triheteromeric NMDARs) for which d-serine might serve as an inverse co-agonist. We will explore how tracing d-serine's origins blends glutamate-receptor biology with glial biology to help provide fresh perspectives on how neurodegeneration might interlink with neuroinflammation to initiate and perpetuate the disease state. Taken together, we envisage the review to deepen our understanding of endogenous d-serine's new role in the brain while also recognizing its therapeutic potential in the treatment of TLE that is oftentimes refractory to medications.
Collapse
Affiliation(s)
- Stephen Beesley
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience Florida State University, 1115 W. Call Street, Tallahassee, FL, 32306-4300, USA
| | - Sanjay S Kumar
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience Florida State University, 1115 W. Call Street, Tallahassee, FL, 32306-4300, USA.
| |
Collapse
|
5
|
Arshad MN, Oppenheimer S, Jeong J, Buyukdemirtas B, Naegele JR. Hippocampal transplants of fetal GABAergic progenitors regulate adult neurogenesis in mice with temporal lobe epilepsy. Neurobiol Dis 2022; 174:105879. [PMID: 36183946 DOI: 10.1016/j.nbd.2022.105879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
GABAergic interneurons play a role in regulating adult neurogenesis within the dentate gyrus (DG) of the hippocampus. Neurogenesis occurs within a stem cell niche in the subgranular zone (SGZ) of the DG. In this niche, populations of neural progenitors give rise to granule cells that migrate radially into the granule cell layer of the DG. Altered neurogenesis in temporal lobe epilepsy (TLE) is linked to a transient increase in the proliferation of new neurons and the abnormal inversion of Type 1 progenitors, resulting in ectopic migration of Type 3 progenitors into the hilus of the DG. These ectopic cells mature into granule cells in the hilus that become hyperexcitable and contribute to the development of spontaneous recurrent seizures. To test whether grafts of GABAergic cells in the DG restore synaptic inhibition, prior work focused on transplanting GABAergic progenitors into the hilus of the DG. This cell-based therapeutic approach was shown to alter the disease phenotype by ameliorating spontaneous seizures in mice with pilocarpine-induced TLE. Prior optogenetic and immunohistochemical studies demonstrated that the transplanted GABAergic interneurons increased levels of synaptic inhibition by establishing inhibitory synaptic contacts with adult-born granule cells, consistent with the observed suppression of seizures. Whether GABAergic progenitor transplantation into the DG ameliorates underlying abnormalities in adult neurogenesis caused by TLE is not known. As a first step to address this question, we compared the effects of GABAergic progenitor transplantation on Type 1, Type 2, and Type 3 progenitors in the stem cell niche using cell type-specific molecular markers in naïve, non-epileptic mice. The progenitor transplantation increased GABAergic interneurons in the DG and led to a significant reduction in Type 2 progenitors and a concomitant increase in Type 3 progenitors. Next, we compared the effects of GABAergic interneuron transplantation in epileptic mice. Transplantation of GABAergic progenitors resulted in reductions in inverted Type 1, Type 2, and hilar ectopic Type 3 cells, concomitant with an increase in the radial migration of Type 3 progenitors into the GCL (Granule Cell Layer). Thus, in mice with Pilocarpine induced TLE, hilar transplants of GABA interneurons may reverse abnormal patterns of adult neurogenesis, an outcome that may ameliorate seizures.
Collapse
Affiliation(s)
- Muhammad N Arshad
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Simon Oppenheimer
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Jaye Jeong
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Bilge Buyukdemirtas
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Janice R Naegele
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| |
Collapse
|
6
|
B. Szabo A, Cretin B, Gérard F, Curot J, J. Barbeau E, Pariente J, Dahan L, Valton L. Sleep: The Tip of the Iceberg in the Bidirectional Link Between Alzheimer's Disease and Epilepsy. Front Neurol 2022; 13:836292. [PMID: 35481265 PMCID: PMC9035794 DOI: 10.3389/fneur.2022.836292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The observation that a pathophysiological link might exist between Alzheimer's disease (AD) and epilepsy dates back to the identification of the first cases of the pathology itself and is now strongly supported by an ever-increasing mountain of literature. An overwhelming majority of data suggests not only a higher prevalence of epilepsy in Alzheimer's disease compared to healthy aging, but also that AD patients with a comorbid epileptic syndrome, even subclinical, have a steeper cognitive decline. Moreover, clinical and preclinical investigations have revealed a marked sleep-related increase in the frequency of epileptic activities. This characteristic might provide clues to the pathophysiological pathways underlying this comorbidity. Furthermore, the preferential sleep-related occurrence of epileptic events opens up the possibility that they might hasten cognitive decline by interfering with the delicately orchestrated synchrony of oscillatory activities implicated in sleep-related memory consolidation. Therefore, we scrutinized the literature for mechanisms that might promote sleep-related epileptic activity in AD and, possibly dementia onset in epilepsy, and we also aimed to determine to what degree and through which processes such events might alter the progression of AD. Finally, we discuss the implications for patient care and try to identify a common basis for methodological considerations for future research and clinical practice.
Collapse
Affiliation(s)
- Anna B. Szabo
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- *Correspondence: Anna B. Szabo
| | - Benjamin Cretin
- Clinical Neuropsychology Unit, Neurology Department, CM2R (Memory Resource and Research Centre), University Hospital of Strasbourg, Strasbourg, France
- CNRS, ICube Laboratory, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), Team IMIS, University of Strasbourg, Strasbourg, France
- CMRR d'Alsace, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Pôle Tête et Cou, Strasbourg, France
| | - Fleur Gérard
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jonathan Curot
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Emmanuel J. Barbeau
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
| | - Jérémie Pariente
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Toulouse NeuroImaging Center (ToNIC), INSERM-University of Toulouse Paul Sabatier, Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Luc Valton
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Luc Valton
| |
Collapse
|
7
|
Moura DMS, de Sales IRP, Brandão JA, Costa MR, Queiroz CM. Disentangling chemical and electrical effects of status epilepticus-induced dentate gyrus abnormalities. Epilepsy Behav 2021; 121:106575. [PMID: 31704249 DOI: 10.1016/j.yebeh.2019.106575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/10/2019] [Accepted: 09/14/2019] [Indexed: 01/06/2023]
Abstract
In rodents, status epilepticus (SE) triggered by chemoconvulsants can differently affect the proliferation and fate of adult-born dentate granule cells (DGCs). It is unknown whether abnormal neurogenesis results from intracellular signaling associated with drug-receptor interaction, paroxysmal activity, or both. To test the contribution of these factors, we systematically compared the effects of kainic acid (KA)- and pilocarpine (PL)-induced SE on the morphology and localization of DGCs generated before or after SE in the ipsi- and contralateral hippocampi of mice. Hippocampal insult was induced by unilateral intrahippocampal (ihpc) administration of KA or PL. We employed conditional doublecortin-dependent expression of the green fluorescent protein (GFP) to label adult-born cells committed to neuronal lineage either one month before (mature DGCs) or seven days after (immature DGCs) SE. Unilateral ihpc administration of KA and PL led to bilateral epileptiform discharges and focal and generalized behavioral seizures. However, drastic granule cell layer (GCL) dispersion occurred only in the ipsilateral side of KA injection, but not in PL-treated animals. Granule cell layer dispersion was accompanied by a significant reduction in neurogenesis after SE in the ipsilateral side of KA-treated animals, while neurogenesis increased in the contralateral side of KA-treated animals and both hippocampi of PL-treated animals. The ratio of ectopic neurons in the ipsilateral hippocampus was higher among immature as compared to mature neurons in the KA model (32.8% vs. 10.0%, respectively), while the occurrence of ectopic neurons in PL-treated animals was lower than 3% among both mature and immature DGCs. Collectively, our results suggest that KA- and PL-induced SE leads to distinct cellular alterations in mature and immature DGCs. We also show different local and secondary effects of KA or PL in the histological organization of the adult DG, suggesting that these unique epilepsy models may be complementary to our understanding of the disease. NEWroscience 2018.
Collapse
|
8
|
Chen L, Wang Y, Chen Z. Adult Neurogenesis in Epileptogenesis: An Update for Preclinical Finding and Potential Clinical Translation. Curr Neuropharmacol 2021; 18:464-484. [PMID: 31744451 PMCID: PMC7457402 DOI: 10.2174/1570159x17666191118142314] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/31/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
Epileptogenesis refers to the process in which a normal brain becomes epileptic, and is characterized by hypersynchronous spontaneous recurrent seizures involving a complex epileptogenic network. Current available pharmacological treatment of epilepsy is generally symptomatic in controlling seizures but is not disease-modifying in epileptogenesis. Cumulative evidence suggests that adult neurogenesis, specifically in the subgranular zone of the hippocampal dentate gyrus, is crucial in epileptogenesis. In this review, we describe the pathological changes that occur in adult neurogenesis in the epileptic brain and how adult neurogenesis is involved in epileptogenesis through different interventions. This is followed by a discussion of some of the molecular signaling pathways involved in regulating adult neurogenesis, which could be potential druggable targets for epileptogenesis. Finally, we provide perspectives on some possible research directions for future studies.
Collapse
Affiliation(s)
- Liying Chen
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Clark LR, Yun S, Acquah NK, Kumar PL, Metheny HE, Paixao RCC, Cohen AS, Eisch AJ. Mild Traumatic Brain Injury Induces Transient, Sequential Increases in Proliferation, Neuroblasts/Immature Neurons, and Cell Survival: A Time Course Study in the Male Mouse Dentate Gyrus. Front Neurosci 2021; 14:612749. [PMID: 33488351 PMCID: PMC7817782 DOI: 10.3389/fnins.2020.612749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are prevalent worldwide. mTBIs can impair hippocampal-based functions such as memory and cause network hyperexcitability of the dentate gyrus (DG), a key entry point to hippocampal circuitry. One candidate for mediating mTBI-induced hippocampal cognitive and physiological dysfunction is injury-induced changes in the process of DG neurogenesis. There are conflicting results on how TBI impacts the process of DG neurogenesis; this is not surprising given that both the neurogenesis process and the post-injury period are dynamic, and that the quantification of neurogenesis varies widely in the literature. Even within the minority of TBI studies focusing specifically on mild injuries, there is disagreement about if and how mTBI changes the process of DG neurogenesis. Here we utilized a clinically relevant rodent model of mTBI (lateral fluid percussion injury, LFPI), gold-standard markers and quantification of the neurogenesis process, and three time points post-injury to generate a comprehensive picture of how mTBI affects adult hippocampal DG neurogenesis. Male C57BL/6J mice (6-8 weeks old) received either sham surgery or mTBI via LFPI. Proliferating cells, neuroblasts/immature neurons, and surviving cells were quantified via stereology in DG subregions (subgranular zone [SGZ], outer granule cell layer [oGCL], molecular layer, and hilus) at short-term (3 days post-injury, dpi), intermediate (7 dpi), and long-term (31 dpi) time points. The data show this model of mTBI induces transient, sequential increases in ipsilateral SGZ/GCL proliferating cells, neuroblasts/immature neurons, and surviving cells which is suggestive of mTBI-induced neurogenesis. In contrast to these ipsilateral hemisphere findings, measures in the contralateral hemisphere were not increased in key neurogenic DG subregions after LFPI. Our work in this mTBI model is in line with most literature on other and more severe models of TBI in showing TBI stimulates the process of DG neurogenesis. However, as our DG data in mTBI provide temporal, subregional, and neurogenesis-stage resolution, these data are important to consider in regard to the functional importance of TBI-induction of the neurogenesis process and future work assessing the potential of replacing and/or repairing DG neurons in the brain after TBI.
Collapse
Affiliation(s)
- Lyles R. Clark
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Nana K. Acquah
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Biological Basis of Behavior Program, University of Pennsylvania, Philadelphia, PA, United States
| | - Priya L. Kumar
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Biomechanical Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Hannah E. Metheny
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
| | - Rikley C. C. Paixao
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
| | - Akivas S. Cohen
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia (CHOP) Research Institute, Philadelphia, PA, United States
- Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Antagonism of Macrophage Migration Inhibitory Factory (MIF) after Traumatic Brain Injury Ameliorates Astrocytosis and Peripheral Lymphocyte Activation and Expansion. Int J Mol Sci 2020; 21:ijms21207448. [PMID: 33050322 PMCID: PMC7589344 DOI: 10.3390/ijms21207448] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) precedes the onset of epilepsy in up to 15–20% of symptomatic epilepsies and up to 5% of all epilepsy. Treatment of acquired epilepsies, including post-traumatic epilepsy (PTE), presents clinical challenges, including frequent resistance to anti-epileptic therapies. Considering that over 1.6 million Americans present with a TBI each year, PTE is an urgent clinical problem. Neuroinflammation is thought to play a major causative role in many of the post-traumatic syndromes, including PTE. Increasing evidence suggests that neuroinflammation facilitates and potentially contributes to seizure induction and propagation. The inflammatory cytokine, macrophage migration inhibitory factor (MIF), is elevated after TBI and higher levels of MIF correlate with worse post-traumatic outcomes. MIF was recently demonstrated to directly alter the firing dynamics of CA1 pyramidal neurons in the hippocampus, a structure critically involved in many types of seizures. We hypothesized that antagonizing MIF after TBI would be anti-inflammatory, anti-neuroinflammatory and neuroprotective. The results show that administering the MIF antagonist ISO1 at 30 min after TBI prevented astrocytosis but was not neuroprotective in the peri-lesion cortex. The results also show that ISO1 inhibited the TBI-induced increase in γδT cells in the gut, and the percent of B cells infiltrating into the brain. The ISO1 treatment also increased this population of B cells in the spleen. These findings are discussed with an eye towards their therapeutic potential for post-traumatic syndromes, including PTE.
Collapse
|
11
|
Hwang Y, Kim HC, Shin EJ. Enhanced neurogenesis is involved in neuroprotection provided by rottlerin against trimethyltin-induced delayed apoptotic neuronal damage. Life Sci 2020; 262:118494. [PMID: 32991881 DOI: 10.1016/j.lfs.2020.118494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/12/2020] [Accepted: 09/20/2020] [Indexed: 12/26/2022]
Abstract
AIMS We here investigated the effect of late- and post-ictal treatment with rottlerin, a polyphenol compound isolated from Mallotus philippinensis, on delayed apoptotic neuronal death induced by trimethyltin (TMT) in mice. MAIN METHODS Male C57BL/6N mice received a single injection of TMT (2.4 mg/kg, i.p.), and mice were treated with rottlerin after a peak time (i.e., 2 d post-TMT) of convulsive behaviors and apoptotic cell death (5.0 mg/kg, i.p. at 3 and 4 d after TMT injection). Object location test and tail suspension test were performed at 5 d after TMT injection. In addition, changes in the expression of apoptotic and neurogenic markers in the dentate gyrus were examined. KEY FINDINGS Late- and post-ictal treatment with rottlerin suppressed delayed neuronal apoptosis in the dentate gyrus, and attenuated memory impairments (as evaluated by object location test) and depression-like behaviors (as evaluated by tail suspension test) at 5 days after TMT injection in mice. In addition, rottlerin enhanced the expression of Sox2 and DCX, and facilitated p-ERK expression in BrdU-incorporated cells in the dentate gyrus of TMT-treated mice. Rottlerin also increased p-Akt expression, and attenuated the increase in the ratio of pro-apoptotic factors/anti-apoptotic factors, and consequent cytosolic cytochrome c release and caspase-3 cleavage. Rottlerin-mediated action was significantly reversed by SL327, an ERK inhibitor. SIGNIFICANCE Our results suggest that late- and post-ictal treatment with rottlerin attenuates TMT-induced delayed neuronal apoptosis in the dentate gyrus of mice via promotion of neurogenesis and inhibition of an on-going apoptotic process through up-regulation of p-ERK.
Collapse
Affiliation(s)
- Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
12
|
Mukherjee S, Arisi GM, Mims K, Hollingsworth G, O'Neil K, Shapiro LA. Neuroinflammatory mechanisms of post-traumatic epilepsy. J Neuroinflammation 2020; 17:193. [PMID: 32552898 PMCID: PMC7301453 DOI: 10.1186/s12974-020-01854-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) occurs in as many as 64-74 million people worldwide each year and often results in one or more post-traumatic syndromes, including depression, cognitive, emotional, and behavioral deficits. TBI can also increase seizure susceptibility, as well as increase the incidence of epilepsy, a phenomenon known as post-traumatic epilepsy (PTE). Injury type and severity appear to partially predict PTE susceptibility. However, a complete mechanistic understanding of risk factors for PTE is incomplete. MAIN BODY From the earliest days of modern neuroscience, to the present day, accumulating evidence supports a significant role for neuroinflammation in the post-traumatic epileptogenic progression. Notably, substantial evidence indicates a role for astrocytes, microglia, chemokines, and cytokines in PTE progression. Although each of these mechanistic components is discussed in separate sections, it is highly likely that it is the totality of cellular and neuroinflammatory interactions that ultimately contribute to the epileptogenic progression following TBI. CONCLUSION This comprehensive review focuses on the neuroinflammatory milieu and explores putative mechanisms involved in the epileptogenic progression from TBI to increased seizure-susceptibility and the development of PTE.
Collapse
Affiliation(s)
- Sanjib Mukherjee
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Gabriel M Arisi
- Department of Physiology, Federal University of Sao Paulo - Escola Paulista de Medicina, Sao Paulo, Brazil.
| | - Kaley Mims
- Texas A&M University, College Station, TX, USA
| | | | | | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA.
| |
Collapse
|
13
|
Sasaki-Takahashi N, Shinohara H, Shioda S, Seki T. The polarity and properties of radial glia-like neural stem cells are altered by seizures with status epilepticus: Study using an improved mouse pilocarpine model of epilepsy. Hippocampus 2020; 30:250-262. [PMID: 32101365 DOI: 10.1002/hipo.23153] [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] [Received: 04/02/2019] [Revised: 08/04/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023]
Abstract
In the adult mouse hippocampus, new neurons are produced by radial glia-like (RGL) neural stem cells in the subgranular zone, which extend their apical processes toward the molecular layer, and express the astrocyte marker glial fibrillary acidic protein, but not the astrocyte marker S100β. In rodent models of epilepsy, adult hippocampal neurogenesis was reported to be increased after acute and mild seizures, but to be decreased by chronic and severe epilepsy. In the present study, we investigated how the severity of seizures affects neurogenesis and RGL neural stem cells in acute stages of epilepsy, using an improved mouse pilocarpine model in which pilocarpine-induced hypothermia was prevented by maintaining body temperature, resulting in a high incidence rate of epileptic seizures and low rate of mortality. In mice that experienced seizures without status epilepticus (SE), the number of proliferating progenitors and immature neurons were significantly increased, whereas no changes were observed in RGL cells. In mice that experienced seizures with SE, the number of proliferating progenitors and immature neurons were unchanged, but the number of RGL cells with an apical process was significantly reduced. Furthermore, the processes of the majority of RGL cells extended inversely toward the hilus, and about half of the aberrant RGL cells expressed S100β. These results suggest that seizures with SE lead to changes in the polarity and properties of RGL neural stem cells, which may direct them toward astrocyte differentiation, resulting in the reduction of neural stem cells producing new granule cells. This also suggests the possibility that cell polarity of RGL stem cells is important for maintaining the stemness of adult neural stem cells.
Collapse
Affiliation(s)
| | - Hiroshi Shinohara
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Seiji Shioda
- Institute for Advanced Bioscience Research, Hoshi University, Tokyo, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
14
|
Targeting Seizure-Induced Neurogenesis in a Clinically Relevant Time Period Leads to Transient But Not Persistent Seizure Reduction. J Neurosci 2019; 39:7019-7028. [PMID: 31308098 DOI: 10.1523/jneurosci.0920-19.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 11/21/2022] Open
Abstract
Mesial temporal lobe epilepsy (mTLE), the most common form of medically refractory epilepsy in adults, is usually associated with hippocampal pathophysiology. Using rodent models of mTLE, many studies including work from our laboratory have shown that new neurons born around the onset of severe acute seizures known as status epilepticus (SE) are crucial for the process of epileptogenesis and targeting seizure-induced neurogenesis either genetically or pharmacologically can impact the frequency of chronic seizures. However, these studies are limited in their clinical relevance as none of them determines the potential of blocking new neurons generated after the epileptogenic insult to alleviate the development of chronic seizures. Therefore, using a pilocarpine-induced SE model of mTLE in mice of either sex, we show that >4 weeks of continuous and concurrent ablation of seizure-induced neurogenesis after SE can reduce the formation of spontaneous recurrent seizures by 65%. We also found that blocking post-SE neurogenesis does not lead to long-term seizure reduction as the effect was observed only transiently for 10 d with >4 weeks of continuous and concurrent ablation of seizure-induced neurogenesis. Thus, these findings provide evidence that seizure-induced neurogenesis when adequately reduced in a clinically relevant time period has the potential to transiently suppress recurrent seizures, but additional mechanisms need to be targeted to permanently prevent epilepsy development.SIGNIFICANCE STATEMENT Consistent with morphological and electrophysiological studies suggesting aberrant adult-generated neurons contribute to epilepsy development, ablation of seizure-induced new neurons at the time of the initial insult reduces the frequency of recurrent seizures. In this study, we show that continuous targeting of post-insult new neurons in a therapeutically relevant time period reduces chronic seizures; however, this effect does not persist suggesting possible additional mechanisms.
Collapse
|
15
|
Shapiro LA. Altered Hippocampal Neurogenesis during the First 7 Days after a Fluid Percussion Traumatic Brain Injury. Cell Transplant 2018; 26:1314-1318. [PMID: 28933222 PMCID: PMC5657729 DOI: 10.1177/0963689717714099] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a devastating disorder causing negative outcomes in millions of people each year. Despite the alarming number of brain injuries and the long-term detrimental outcomes that can be associated with TBI, treatment options are lacking. Extensive investigation is underway, in hopes of identifying effective treatment strategies. Among the most state-of-the-art strategies is cell replacement therapy. TBI is a seemingly good candidate for cell replacement studies because there is often loss of neurons. However, translation of this therapy has not yet been successful. It is possible that a better understanding of endogenous neurogenic mechanisms after TBI could lead to more efficacious study designs using exogenous cell replacement strategies. Therefore, this study was designed to examine the number and migration of immature neurons at 1 and 7 d after a fluid percussion TBI. The results show that the number of immature neurons increases from 7 d after a fluid percussion injury (FPI), and there is ectopic migration of doublecortin (DCX+) immature neurons into the hilar region of the dentate gyrus. These results add important data to the current understanding of the endogenous neurogenic niche after TBI. Follow-up studies are needed to better understand the functional significance of elevated neurogenesis and aberrant migration into the hilus.
Collapse
Affiliation(s)
- Lee A Shapiro
- 1 Department of Surgery, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
16
|
Magagna-Poveda A, Moretto JN, Scharfman HE. Increased gyrification and aberrant adult neurogenesis of the dentate gyrus in adult rats. Brain Struct Funct 2017; 222:4219-4237. [PMID: 28656372 PMCID: PMC5909844 DOI: 10.1007/s00429-017-1457-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023]
Abstract
A remarkable example of maladaptive plasticity is the development of epilepsy after a brain insult or injury to a normal animal or human. A structure that is considered central to the development of this type of epilepsy is the dentate gyrus (DG), because it is normally a relatively inhibited structure and its quiescence is thought to reduce hippocampal seizure activity. This characteristic of the DG is also considered to be important for normal hippocampal-dependent cognitive functions. It has been suggested that the brain insults which cause epilepsy do so because they cause the DG to be more easily activated. One type of brain insult that is commonly used is induction of severe seizures (status epilepticus; SE) by systemic injection of a convulsant drug. Here we describe an alteration in the DG after this type of experimental SE that may contribute to chronic seizures that has not been described before: large folds or gyri that develop in the DG by 1 month after SE. Large gyri appeared to increase network excitability because epileptiform discharges recorded in hippocampal slices after SE were longer in duration when recorded inside gyri relative to locations outside gyri. Large gyri may also increase excitability because immature adult-born neurons accumulated at the base of gyri with time after SE, and previous studies have suggested that abnormalities in adult-born DG neurons promote seizures after SE. In summary, large gyri after SE are a common finding in adult rats, show increased excitability, and are associated with the development of an abnormal spatial distribution of adult-born neurons. Together these alterations may contribute to chronic seizures and associated cognitive comorbidities after SE.
Collapse
Affiliation(s)
- Alejandra Magagna-Poveda
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, 140 Old Orangeburg Rd. Bldg. 35, Orangeburg, NY, 10962, USA
| | - Jillian N Moretto
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, 140 Old Orangeburg Rd. Bldg. 35, Orangeburg, NY, 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, 140 Old Orangeburg Rd. Bldg. 35, Orangeburg, NY, 10962, USA.
- Department of Child and Adolescent Psychiatry, New York University Langone Medical Center, One Park Ave., New York, NY, 10016, USA.
- Department of Physiology and Neuroscience, New York University Langone Medical Center, One Park Ave., New York, NY, 10016, USA.
- Department of Psychiatry, New York University Langone Medical Center, One Park Ave., New York, NY, 10016, USA.
| |
Collapse
|
17
|
Woitke F, Ceanga M, Rudolph M, Niv F, Witte OW, Redecker C, Kunze A, Keiner S. Adult hippocampal neurogenesis poststroke: More new granule cells but aberrant morphology and impaired spatial memory. PLoS One 2017; 12:e0183463. [PMID: 28910298 PMCID: PMC5598932 DOI: 10.1371/journal.pone.0183463] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/04/2017] [Indexed: 01/29/2023] Open
Abstract
Stroke significantly stimulates neurogenesis in the adult dentate gyrus, though the functional role of this postlesional response is mostly unclear. Recent findings suggest that newborn neurons generated in the context of stroke may fail to correctly integrate into pre-existing networks. We hypothesized that increased neurogenesis in the dentate gyrus following stroke is associated with aberrant neurogenesis and impairment of hippocampus-dependent memory. To address these questions we used the middle cerebral artery occlusion model (MCAO) in mice. Animals were housed either under standard conditions or with free access to running wheels. Newborn granule cells were labelled with the thymidine analoque EdU and retroviral vectors. To assess memory performance, we employed a modified version of the Morris water maze (MWM) allowing differentiation between hippocampus dependent and independent learning strategies. Newborn neurons were morphologically analyzed using confocal microscopy and Neurolucida system at 7 weeks. We found that neurogenesis was significantly increased following MCAO. Animals with MCAO needed more time to localize the platform and employed less hippocampus-dependent search strategies in MWM versus controls. Confocal studies revealed an aberrant cell morphology with basal dendrites and an ectopic location (e.g. hilus) of new granule cells born in the ischemic brain. Running increased the number of new neurons but also enhanced aberrant neurogenesis. Running, did not improve the general performance in the MWM but slightly promoted the application of precise spatial search strategies. In conclusion, ischemic insults cause hippocampal-dependent memory deficits which are associated with aberrant neurogenesis in the dentate gyrus indicating ischemia-induced maladaptive plasticity in the hippocampus.
Collapse
Affiliation(s)
- Florus Woitke
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Mihai Ceanga
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Max Rudolph
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Fanny Niv
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Otto W. Witte
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Christoph Redecker
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| | - Albrecht Kunze
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
- * E-mail:
| | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, Jena, Germany
| |
Collapse
|
18
|
Cipriani R, Chara JC, Rodríguez-Antigüedad A, Matute C. Effects of FTY720 on brain neurogenic niches in vitro and after kainic acid-induced injury. J Neuroinflammation 2017; 14:147. [PMID: 28738875 PMCID: PMC5525223 DOI: 10.1186/s12974-017-0922-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/14/2017] [Indexed: 01/28/2023] Open
Abstract
Background FTY720 (fingolimod, Gilenya™) is an oral, blood-brain barrier (BBB)-passing drug approved as immunomodulatory treatment for relapsing-remitting form of the multiple sclerosis (MS). In addition, FTY720 exerts several effects in the central nervous system (CNS), ranging from neuroprotection to reduction of neuroinflammation. However, the neurogenic and oligodendrogenic potential of FTY720 has been poorly investigated. In this study, we assessed the effect of FTY720 on the production of new neurons and oligodendrocytes from neural stem/precursor cells both in vitro and in vivo. Methods Neural stem cells (NSCs) derived from the young rat subventricular zone (SVZ) were exposed to FTY720 (10, 100 nM), and their differentiation into neurons and oligodendrocytes was measured using immunofluorescence for anti-β-III tubulin or CNPase (2′,3′-cyclic nucleotide 3′-phosphodiesterase) as markers of mature neurons or oligodendrocytes, respectively. In addition, intracerebroventricular (icv) administration of kainic acid (KA; 0.5 μg/2 μl) in Sprague-Dawley rats was used as an in vivo model of neuronal death and inflammation. FTY720 was applied icv (1 μg/2 μl), together with KA, plus intraperitoneally (ip; 1 mg/kg) 24 h before, and daily, until sacrifice 8 days after KA injection. To visualize cell proliferation in the hippocampus and in white matter regions, rats were administered 5-bromo-2-deoxyuridine (BrdU) 100 mg/kg, ip injected every 2 days. Immunohistochemical analyses were performed on rat brain slices to measure the production of new neuronal precursors (doublecortin/DCX+ cells) and new oligodendrocytes precursors (proteoglycan/NG2+ cells). Results In this study, we observed that FTY720 increased postnatal NSCs differentiation into both neurons and oligodendrocytes in vitro. In turn, in adult animals, FTY720 enhanced the percentage of BrdU+ cells coexpressing DCX marker, both in basal (FTY720 alone) and in neurodegenerative (FTY720 + KA) conditions. However, FTY720 had only a partial effect on proliferation and differentiation of oligodendrocyte progenitor cell (OPC) population in vivo. Conclusions FTY720 promotes neurogenesis and oligodendrogenesis in vitro under basal conditions. In addition, it increases the generation of neuroblasts and oligodendrocytes after excitotoxic brain injury. This suggests that FTY720 has the potential to activate the neurogenic niche and thus favour tissue repair after lesion. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0922-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raffaela Cipriani
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain.
| | - Juan Carlos Chara
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain
| | | | - Carlos Matute
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), E-48940, Leioa, Spain
| |
Collapse
|
19
|
Uemori T, Toda K, Seki T. Seizure severity-dependent selective vulnerability of the granule cell layer and aberrant neurogenesis in the rat hippocampus. Hippocampus 2017; 27:1054-1068. [PMID: 28608989 DOI: 10.1002/hipo.22752] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/20/2022]
Abstract
The pilocarpine-induced status epilepticus rodent model has been commonly used to analyze the mechanisms of human temporal lobe epilepsy. Recent studies using this model have demonstrated that epileptic seizures lead to increased adult neurogenesis of the dentate granule cells, and cause abnormal cellular organization in dentate neuronal circuits. In this study, we examined these structural changes in rats with seizures of varying severity. In rats with frequent severe seizures, we found a clear loss of Prox1 and NeuN expression in the dentate granule cell layer (GCL), which was confined mainly to the suprapyramidal blade of the GCL at the septal and middle regions of the septotemporal axis of the hippocampus. In the damaged suprapyramidal region, the number of immature neurons in the subgranular zone was markedly reduced. In contrast, in rats with less frequent severe seizures, there was almost no loss of Prox1 and NeuN expression, and the number of immature neurons was increased. In rats with no or slight loss of Prox1 expression in the GCL, ectopic immature neurons were detected in the molecular layer of the suprapyramidal blade in addition to the hilus, and formed chainlike aggregated structures along the blood vessels up to the hippocampal fissure, suggesting that newly generated neurons migrate at least partially along blood vessels to the hippocampal fissure. These results suggest that seizures of different severity cause different effects on GCL damage, neurogenesis, and the migration of new neurons, and that these structural changes are selective to subdivisions of the GCL and the septotemporal axis of the hippocampus.
Collapse
Affiliation(s)
- Takeshi Uemori
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Keiko Toda
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
20
|
The Effects of Amiloride on Seizure Activity, Cognitive Deficits and Seizure-Induced Neurogenesis in a Novel Rat Model of Febrile Seizures. Neurochem Res 2015; 41:933-42. [DOI: 10.1007/s11064-015-1777-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/02/2015] [Accepted: 11/14/2015] [Indexed: 12/14/2022]
|
21
|
Singh SP, LaSarge CL, An A, McAuliffe JJ, Danzer SC. Clonal Analysis of Newborn Hippocampal Dentate Granule Cell Proliferation and Development in Temporal Lobe Epilepsy. eNeuro 2015; 2:ENEURO.0087-15.2015. [PMID: 26756038 PMCID: PMC4706641 DOI: 10.1523/eneuro.0087-15.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 11/21/2022] Open
Abstract
Hippocampal dentate granule cells are among the few neuronal cell types generated throughout adult life in mammals. In the normal brain, new granule cells are generated from progenitors in the subgranular zone and integrate in a typical fashion. During the development of epilepsy, granule cell integration is profoundly altered. The new cells migrate to ectopic locations and develop misoriented "basal" dendrites. Although it has been established that these abnormal cells are newly generated, it is not known whether they arise ubiquitously throughout the progenitor cell pool or are derived from a smaller number of "bad actor" progenitors. To explore this question, we conducted a clonal analysis study in mice expressing the Brainbow fluorescent protein reporter construct in dentate granule cell progenitors. Mice were examined 2 months after pilocarpine-induced status epilepticus, a treatment that leads to the development of epilepsy. Brain sections were rendered translucent so that entire hippocampi could be reconstructed and all fluorescently labeled cells identified. Our findings reveal that a small number of progenitors produce the majority of ectopic cells following status epilepticus, indicating that either the affected progenitors or their local microenvironments have become pathological. By contrast, granule cells with "basal" dendrites were equally distributed among clonal groups. This indicates that these progenitors can produce normal cells and suggests that global factors sporadically disrupt the dendritic development of some new cells. Together, these findings strongly predict that distinct mechanisms regulate different aspects of granule cell pathology in epilepsy.
Collapse
Affiliation(s)
- Shatrunjai P. Singh
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229
- Molecular and Developmental Biology Program, University of Cincinnati, Cincinnati, Ohio 45237
| | - Candi L. LaSarge
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229
| | - Amen An
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229
- Department of Neuroscience, McMicken College of Arts and Sciences, University of Cincinnati, Cincinnati, Ohio 45221
| | - John J. McAuliffe
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229
| | - Steve C. Danzer
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229
- Molecular and Developmental Biology Program, University of Cincinnati, Cincinnati, Ohio 45237
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, Ohio 45267
| |
Collapse
|
22
|
Ko AR, Hyun HW, Min SJ, Kim JE, Kang TC. Endothelin-1 induces LIMK2-mediated programmed necrotic neuronal death independent of NOS activity. Mol Brain 2015; 8:58. [PMID: 26438559 PMCID: PMC4595180 DOI: 10.1186/s13041-015-0149-3] [Citation(s) in RCA: 20] [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: 07/02/2015] [Accepted: 09/18/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Recently, we have reported that LIM kinase 2 (LIMK2) involves programmed necrotic neuronal deaths induced by aberrant cyclin D1 expression following status epilepticus (SE). Up-regulation of LIMK2 expression induces neuronal necrosis by impairment of dynamin-related protein 1 (DRP1)-mediated mitochondrial fission. However, we could not elucidate the upstream effecter for LIMK2-mediated neuronal death. Thus, we investigated the role of endothelin-1 (ET-1) in LIMK2-mediated neuronal necrosis, since ET-1 involves neuronal death via various pathways. RESULTS Following SE, ET-1 concentration and its mRNA were significantly increased in the hippocampus with up-regulation of ETB receptor expression. BQ788 (an ETB receptor antagonist) effectively attenuated SE-induced neuronal damage as well as reduction in LIMK2 mRNA/protein expression. In addition, BQ788 alleviated up-regulation of Rho kinase 1 (ROCK1) expression and impairment of DRP1-mediated mitochondrial fission in CA1 neurons following SE. BQ788 also attenuated neuronal death and up-regulation of LIMK2 expression induced by exogenous ET-1 injection. CONCLUSION These findings suggest that ET-1 may be one of the upstream effectors for programmed neuronal necrosis through abnormal LIMK2 over-expression by ROCK1.
Collapse
Affiliation(s)
- Ah-Reum Ko
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Hye-Won Hyun
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Su-Ji Min
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea
| | - Ji-Eun Kim
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea.
| | - Tae-Cheon Kang
- Department of Anatomy & Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, 200-702, South Korea.
| |
Collapse
|
23
|
Functional Integration of Adult-Born Hippocampal Neurons after Traumatic Brain Injury(1,2,3). eNeuro 2015; 2:eN-NWR-0056-15. [PMID: 26478908 PMCID: PMC4603252 DOI: 10.1523/eneuro.0056-15.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 01/31/2023] Open
Abstract
Traumatic brain injury (TBI) increases hippocampal neurogenesis, which may contribute to cognitive recovery after injury. However, it is unknown whether TBI-induced adult-born neurons mature normally and functionally integrate into the hippocampal network. We assessed the generation, morphology, and synaptic integration of new hippocampal neurons after a controlled cortical impact (CCI) injury model of TBI. To label TBI-induced newborn neurons, we used 2-month-old POMC-EGFP mice, which transiently and specifically express EGFP in immature hippocampal neurons, and doublecortin-CreERT2 transgenic mice crossed with Rosa26-CAG-tdTomato reporter mice, to permanently pulse-label a cohort of adult-born hippocampal neurons. TBI increased the generation, outward migration, and dendritic complexity of neurons born during post-traumatic neurogenesis. Cells born after TBI had profound alterations in their dendritic structure, with increased dendritic branching proximal to the soma and widely splayed dendritic branches. These changes were apparent during early dendritic outgrowth and persisted as these cells matured. Whole-cell recordings from neurons generated during post-traumatic neurogenesis demonstrate that they are excitable and functionally integrate into the hippocampal circuit. However, despite their dramatic morphologic abnormalities, we found no differences in the rate of their electrophysiological maturation, or their overall degree of synaptic integration when compared to age-matched adult-born cells from sham mice. Our results suggest that cells born after TBI participate in information processing, and receive an apparently normal balance of excitatory and inhibitory inputs. However, TBI-induced changes in their anatomic localization and dendritic projection patterns could result in maladaptive network properties.
Collapse
|
24
|
Scharfman HE, Bernstein HL. Potential implications of a monosynaptic pathway from mossy cells to adult-born granule cells of the dentate gyrus. Front Syst Neurosci 2015; 9:112. [PMID: 26347618 PMCID: PMC4541026 DOI: 10.3389/fnsys.2015.00112] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
The dentate gyrus (DG) is important to many aspects of hippocampal function, but there are many aspects of the DG that are incompletely understood. One example is the role of mossy cells (MCs), a major DG cell type that is glutamatergic and innervates the primary output cells of the DG, the granule cells (GCs). MCs innervate the GCs as well as local circuit neurons that make GABAergic synapses on GCs, so the net effect of MCs on GCs – and therefore the output of the DG – is unclear. Here we first review fundamental information about MCs and the current hypotheses for their role in the normal DG and in diseases that involve the DG. Then we review previously published data which suggest that MCs are a source of input to a subset of GCs that are born in adulthood (adult-born GCs). In addition, we discuss the evidence that adult-born GCs may support the normal inhibitory ‘gate’ functions of the DG, where the GCs are a filter or gate for information from the entorhinal cortical input to area CA3. The implications are then discussed in the context of seizures and temporal lobe epilepsy (TLE). In TLE, it has been suggested that the DG inhibitory gate is weak or broken and MC loss leads to insufficient activation of inhibitory neurons, causing hyperexcitability. That idea was called the “dormant basket cell hypothesis.” Recent data suggest that loss of normal adult-born GCs may also cause disinhibition, and seizure susceptibility. Therefore, we propose a reconsideration of the dormant basket cell hypothesis with an intervening adult-born GC between the MC and basket cell and call this hypothesis the “dormant immature granule cell hypothesis.”
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan Kline Institute for Psychiatric Research, Orangeburg NY, USA ; New York University Langone Medical Center, New York NY, USA
| | - Hannah L Bernstein
- The Nathan Kline Institute for Psychiatric Research, Orangeburg NY, USA ; New York University Langone Medical Center, New York NY, USA
| |
Collapse
|
25
|
Kim JY, Ko AR, Hyun HW, Kang TC. ETB receptor-mediated MMP-9 activation induces vasogenic edema via ZO-1 protein degradation following status epilepticus. Neuroscience 2015; 304:355-67. [PMID: 26232046 DOI: 10.1016/j.neuroscience.2015.07.065] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/08/2015] [Accepted: 07/24/2015] [Indexed: 01/08/2023]
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells with specialized tight junctions (TJs) lining the blood vessels and astroglial endfeet surrounding the blood vessels. Although BBB disruption during brain insults leads to vasogenic edema as one of the primary steps in the epileptogenic process, little is known about the molecular and physiological events concerning vasogenic edema formation. In the present study, status epilepticus (SE) changed the expressions and subcellular localizations of TJ proteins (claudin-5, occludin and zonula occludens-1 (ZO-1)) in endothelial cells of the rat piriform cortex. Among TJ proteins, the alteration in ZO-1 expression was relevant to endothelin B (ETB) receptor-mediated endothelial nitric oxide synthase (eNOS) activation, which increased matrix metalloproteinase-9 (MMP-9) activity. Indeed, BQ788 (an ETB receptor antagonist) effectively attenuated SE-induced vasogenic edema by inhibiting eNOS-mediated MMP-9 activation and ZO-1 protein degradation in endothelial cells, although astroglial endfeet were detached from endothelial cells. Therefore, we suggest that SE-induced ETB receptor/eNOS-mediated MMP-9 activation may lead to impairments of endothelial cell function via TJ protein degradation, which are involved in vasogenic edema formation independent of perivascular astroglial functions.
Collapse
Affiliation(s)
- J Y Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - A-R Ko
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - H-W Hyun
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | - T-C Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea.
| |
Collapse
|
26
|
Ko AR, Kim JY, Hyun HW, Kim JE. Endothelial NOS activation induces the blood-brain barrier disruption via ER stress following status epilepticus. Brain Res 2015; 1622:163-73. [PMID: 26115585 DOI: 10.1016/j.brainres.2015.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 11/19/2022]
Abstract
The blood-brain barrier (BBB) maintains the unique brain microenvironment, which is separated from the systemic circulating system. Since the endoplasmic reticulum (ER) is an important cell organelle that is responsible for protein synthesis, the correct folding and sorting of proteins contributing to cell survivals, ER stress is a potential cause of cell damage in various diseases. Therefore, it would be worthy to explore the the relationship between the ER stress and BBB disruption during vasogenic edema formation induced by epileptogenic insults. In the present study, we investigated the roles of ER stress in vasogenic edema and its related events in rat epilepsy models provoked by pilocarpine-induced status epilepticus (SE). SE-induced eNOS activation induces BBB breakdown via up-regulation of GRP78 expression and dysfunction of SMI-71 (an endothelial BBB marker) in the piriform cortex (PC). In addition, caveolin-1 peptide (an eNOS inhibitor) effectively attenuated GRP78 expression and down-regulation of SMI-71. Taken together, our findings suggest that eNOS-mediated ER stress may participate in SE-induced vasogenic edema formation. Therefore, the modulation of ER stress may be a considerable strategy for therapy in impairments of endothelial cell function.
Collapse
Affiliation(s)
- Ah-Reum Ko
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Ji Yang Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Hye-Won Hyun
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, Kangwon-Do 200702, South Korea.
| |
Collapse
|
27
|
Iyengar SS, LaFrancois JJ, Friedman D, Drew LJ, Denny CA, Burghardt NS, Wu MV, Hsieh J, Hen R, Scharfman HE. Suppression of adult neurogenesis increases the acute effects of kainic acid. Exp Neurol 2015; 264:135-49. [PMID: 25476494 PMCID: PMC4800819 DOI: 10.1016/j.expneurol.2014.11.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 11/10/2014] [Accepted: 11/20/2014] [Indexed: 01/17/2023]
Abstract
Adult neurogenesis, the generation of new neurons in the adult brain, occurs in the hippocampal dentate gyrus (DG) and the olfactory bulb (OB) of all mammals, but the functions of these new neurons are not entirely clear. Originally, adult-born neurons were considered to have excitatory effects on the DG network, but recent studies suggest a net inhibitory effect. Therefore, we hypothesized that selective removal of newborn neurons would lead to increased susceptibility to the effects of a convulsant. This hypothesis was tested by evaluating the response to the chemoconvulsant kainic acid (KA) in mice with reduced adult neurogenesis, produced either by focal X-irradiation of the DG, or by pharmacogenetic deletion of dividing radial glial precursors. In the first 4 hrs after KA administration, when mice have the most robust seizures, mice with reduced adult neurogenesis had more severe convulsive seizures, exhibited either as a decreased latency to the first convulsive seizure, greater number of convulsive seizures, or longer convulsive seizures. Nonconvulsive seizures did not appear to change or they decreased. Four-21 hrs after KA injection, mice with reduced adult neurogenesis showed more interictal spikes (IIS) and delayed seizures than controls. Effects were greater when the anticonvulsant ethosuximide was injected 30 min prior to KA administration; ethosuximide allows forebrain seizure activity to be more easily examined in mice by suppressing seizures dominated by the brainstem. These data support the hypothesis that reduction of adult-born neurons increases the susceptibility of the brain to effects of KA.
Collapse
Affiliation(s)
- Sloka S Iyengar
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - John J LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Daniel Friedman
- Department of Neurology, New York University Langone Medical Center, New York, NY 10016
| | - Liam J Drew
- WIBR, University College of London, London, UK WC1E 6BT
| | - Christine A Denny
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY 10032
| | - Nesha S Burghardt
- Department of Psychology, Hunter College, City University of New York, New York, NY 10065
| | - Melody V Wu
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY 10032
| | - Jenny Hsieh
- Department of Molecular Neurobiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - René Hen
- Department of Psychiatry, Columbia University, College of Physicians and Surgeons, New York, NY 10032; Department of Molecular Neurobiology, University of Texas Southwestern Medical Center, Dallas, TX 75390; New York State Psychiatric Institute, New York, NY 10032
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962; Departments of Child & Adolescent Psychiatry, Physiology & Neuroscience, and Psychiatry, New York University Langone Medical Center, New York, NY 10016.
| |
Collapse
|
28
|
In vivo effects of bumetanide at brain concentrations incompatible with NKCC1 inhibition on newborn DGC structure and spontaneous EEG seizures following hypoxia-induced neonatal seizures. Neuroscience 2014; 286:203-15. [PMID: 25463517 DOI: 10.1016/j.neuroscience.2014.11.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 12/21/2022]
Abstract
Neonatal seizures caused by perinatal asphyxia and hypoxic-ischemic encephalopathy can be refractory to conventional anticonvulsants. This may be due to the depolarizing effects of gamma-aminobutyric acid (GABA) achieved by the activity of the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1). The aim of this study is to evaluate the long-term effects of bumetanide, a NKCC1 inhibitor, on hippocampal neurogenesis and seizure susceptibility in hypoxia-induced neonatal seizure model. Wistar rats were subjected to hypoxia-induced neonatal seizures at postnatal day 10 (P10). Following acute seizures, the rats were treated with intraperitoneal injection (i.p.) of bumetanide at a dose of 0.5mg/kg for 3 weeks. In later adulthood, hypoxia-induced seizures increased the number of newborn dentate gyrus cells (DGCs), promoted mossy fiber sprouting (MFS) and reduced the apical dendritic complexity of newborn DGCs 1 month after the insults. In addition, these seizures resulted in long-lasting consequences, such as spontaneous electroencephalography (EEG) seizures, though spatial learning impairments were not seen. Bumetanide treatments significantly enhanced cell proliferation and dendritic development of newborn DGCs after neonatal seizures, accompanied by the decreased seizure activity. However, systemic administration of bumetanide resulted in much lower brain concentrations, and was incompatible with NKCC1 inhibition in blood-brain barrier (BBB)-protected brain tissue. Our results suggested that bumetanide might have long-term effects in suppressing seizure activity, and altering the neurogenesis after neonatal seizures. These effects of bumetanide may be mediated by the targets outside the BBB-protected central nerve system (CNS) or CNS-located target(s) other than NKCC1.
Collapse
|
29
|
Scharfman HE, Brooks-Kayal AR. Is plasticity of GABAergic mechanisms relevant to epileptogenesis? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:133-50. [PMID: 25012373 DOI: 10.1007/978-94-017-8914-1_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Numerous changes in GABAergic neurons, receptors, and inhibitory mechanisms have been described in temporal lobe epilepsy (TLE), either in humans or in animal models. Nevertheless, there remains a common assumption that epilepsy can be explained by simply an insufficiency of GABAergic inhibition. Alternatively, investigators have suggested that there is hyperinhibition that masks an underlying hyperexcitability. Here we examine the status epilepticus (SE) models of TLE and focus on the dentate gyrus of the hippocampus, where a great deal of data have been collected. The types of GABAergic neurons and GABAA receptors are summarized under normal conditions and after SE. The role of GABA in development and in adult neurogenesis is discussed. We suggest that instead of "too little or too much" GABA there is a complexity of changes after SE that makes the emergence of chronic seizures (epileptogenesis) difficult to understand mechanistically, and difficult to treat. We also suggest that this complexity arises, at least in part, because of the remarkable plasticity of GABAergic neurons and GABAA receptors in response to insult or injury.
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA,
| | | |
Collapse
|
30
|
Caspase 3 involves in neuroplasticity, microglial activation and neurogenesis in the mice hippocampus after intracerebral injection of kainic acid. J Biomed Sci 2013; 20:90. [PMID: 24313976 PMCID: PMC4028745 DOI: 10.1186/1423-0127-20-90] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/02/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The roles of caspase 3 on the kainic acid-mediated neurodegeneration, dendritic plasticity alteration, neurogenesis, microglial activation and gliosis are not fully understood. Here, we investigate hippocampal changes using a mouse model that receive a single kainic acid-intracerebral ventricle injection. The effects of caspase 3 inhibition on these changes were detected during a period of 1 to 7 days post kainic acid injection. RESULT Neurodegeneration was assessed by Fluoro-Jade B staining and neuronal nuclei protein (NeuN) immunostaining. Neurogenesis, gliosis, neuritic plasticity alteration and caspase 3 activation were examined using immunohistochemistry. Dendritic plasticity, cleavvage-dependent activation of calcineurin A and glial fibrillary acidic protein cleavage were analyzed by immunoblotting. We found that kainic acid not only induced neurodegeneration but also arouse several caspase 3-mediated molecular and cellular changes including dendritic plasticity, neurogenesis, and gliosis. The acute caspase 3 activation occurred in pyramidal neurons as well as in hilar interneurons. The delayed caspase 3 activation occurred in astrocytes. The co-injection of caspase 3 inhibitor did not rescue kainic acid-mediated neurodegeneration but seriously and reversibly disturb the structural integrity of axon and dendrite. The kainic acid-induced events include microglia activation, the proliferation of radial glial cells, neurogenesis, and calcineurin A cleavage were significantly inhibited by the co-injection of caspase 3 inhibitor, suggesting the direct involvement of caspase 3 in these events. Alternatively, the kainic acid-mediated astrogliosis is not caspase 3-dependent, although caspase 3 cleavage of glial fibrillary acidic protein occurred. CONCLUSIONS Our results provide the first direct evidence of a causal role of caspase 3 activation in the cellular changes during kainic acid-mediated excitotoxicity. These findings may highlight novel pharmacological strategies to arrest disease progression and control seizures that are refractory to classical anticonvulsant treatment.
Collapse
|
31
|
Russmann V, Seeger N, Zellinger C, Hadamitzky M, Pankratova S, Wendt H, Bock E, Berezin V, Potschka H. The CNTF-derived peptide mimetic Cintrofin attenuates spatial-learning deficits in a rat post-status epilepticus model. Neurosci Lett 2013; 556:170-5. [DOI: 10.1016/j.neulet.2013.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/10/2013] [Accepted: 10/01/2013] [Indexed: 12/24/2022]
|
32
|
Kim JE, Ryu HJ, Kang TC. Status epilepticus induces vasogenic edema via tumor necrosis factor-α/ endothelin-1-mediated two different pathways. PLoS One 2013; 8:e74458. [PMID: 24040253 PMCID: PMC3764062 DOI: 10.1371/journal.pone.0074458] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 07/31/2013] [Indexed: 12/11/2022] Open
Abstract
Status epilepticus (SE) induces vasogenic edema in the piriform cortex with disruptions of the blood-brain barrier (BBB). However, the mechanisms of vasogenic edema formation following SE are still unknown. Here we investigated the endothelin B (ETB) receptor-mediated pathway of SE-induced vasogenic edema. Following SE, the release of tumor necrosis factor-α (TNF-α) stimulated endothelin-1 (ET-1) release and expression in neurons and endothelial cells. In addition, TNF-α-induced ET-1 increased BBB permeability via ETB receptor-mediated endothelial nitric oxide synthase (eNOS) activation in endothelial cells. ETB receptor activation also increased intracellular reactive oxygen species by NADPH oxidase production in astrocytes. These findings suggest that SE results in BBB dysfunctions via endothelial-astroglial interactions through the TNF-α-ET-1-eNOS/NADPH oxidase pathway, and that these ETB receptor-mediated interactions may be an effective therapeutic strategy for vasogenic edema in various neurological diseases.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
| | - Hea Jin Ryu
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, Kangwon-Do, Republic of Korea
- * E-mail:
| |
Collapse
|
33
|
The influence of ectopic migration of granule cells into the hilus on dentate gyrus-CA3 function. PLoS One 2013; 8:e68208. [PMID: 23840835 PMCID: PMC3695928 DOI: 10.1371/journal.pone.0068208] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/27/2013] [Indexed: 11/29/2022] Open
Abstract
Postnatal neurogenesis of granule cells (GCs) in the dentate gyrus (DG) produces GCs that normally migrate from the subgranular zone to the GC layer. However, GCs can mismigrate into the hilus, the opposite direction. Previous descriptions of these hilar ectopic GCs (hEGCs) suggest that they are rare unless there are severe seizures. However, it is not clear if severe seizures are required, and it also is unclear if severe seizures are responsible for the abnormalities of hEGCs, which include atypical dendrites and electrophysiological properties. Here we show that large numbers of hEGCs develop in a transgenic mouse without severe seizures. The mice have a deletion of BAX, which normally regulates apoptosis. Surprisingly, we show that hEGCs in the BAX-/- mouse have similar abnormalities as hEGCs that arise after severe seizures. We next asked if there are selective effects of hEGCs, i.e., whether a robust population of hEGCs would have any effect on the DG if they were induced without severe seizures. Indeed, this appears to be true, because it has been reported that BAX-/- mice have defects in a behavior that tests pattern separation, which depends on the DG. However, inferring functional effects of hEGCs is difficult in mice with a constitutive BAX deletion because there is decreased apoptosis in and outside the DG. Therefore, a computational model of the normal DG and hippocampal subfield CA3 was used. Adding a small population of hEGCs (5% of all GCs), with characteristics defined empirically, was sufficient to disrupt a simulation of pattern separation and completion. Modeling results also showed that effects of hEGCs were due primarily to “backprojections” of CA3 pyramidal cell axons to the hilus. The results suggest that hEGCs can develop for diverse reasons, do not depend on severe seizures, and a small population of hEGCs may impair DG-dependent function.
Collapse
|
34
|
Licko T, Seeger N, Zellinger C, Russmann V, Matagne A, Potschka H. Lacosamide treatment following status epilepticus attenuates neuronal cell loss and alterations in hippocampal neurogenesis in a rat electrical status epilepticus model. Epilepsia 2013; 54:1176-85. [PMID: 23614482 DOI: 10.1111/epi.12196] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/22/2022]
Abstract
PURPOSE The antiepileptic drug, lacosamide, exerts its therapeutic activity by enhancing slow inactivation of voltage-gated sodium channels. Because putative preventive or disease-modifying effects of drugs may affect epileptogenesis, intrinsic severity, and comorbidities, it is of particular interest to assess the effect of lacosamide on the development of epilepsy and associated cellular alterations. METHODS The effect of lacosamide was evaluated in an electrical rat status epilepticus (SE) model with a 24-day treatment phase following induction of SE. The impact of lacosamide on the development of spontaneous seizures based on continuous video-electroencephalography (EEG) monitoring, as well as the impact on neuronal cell loss and alterations in hippocampal neurogenesis, was assessed. KEY FINDINGS Neither low-dose nor high-dose lacosamide affected the development of spontaneous seizures. A dose-dependent neuroprotective effect of lacosamide with significant reduction of neuronal cell loss was observed in the hippocampal CA1 region, as well as in the piriform cortex. In addition, lacosamide attenuated the impact of SE on the rate of hippocampal cell neurogenesis. Moreover, lacosamide prevented a significant rise in the number of persistent basal dendrites. SIGNIFICANCE Our data do not support an antiepileptogenic effect of lacosamide. However, because lacosamide reduced SE-associated cellular alterations, it would be of interest to determine whether these effects indicate a putative disease-modifying effect of lacosamide in future studies.
Collapse
Affiliation(s)
- Thomas Licko
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilian-University, Munich, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Chin J, Scharfman HE. Shared cognitive and behavioral impairments in epilepsy and Alzheimer's disease and potential underlying mechanisms. Epilepsy Behav 2013; 26:343-51. [PMID: 23321057 PMCID: PMC3924321 DOI: 10.1016/j.yebeh.2012.11.040] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 11/17/2012] [Indexed: 01/27/2023]
Abstract
Seizures in patients with Alzheimer's disease (AD) have been examined by many investigators over the last several decades, and there are diverse opinions about their potential relevance to AD pathophysiology. Some studies suggest that seizures appear to be a fairly uncommon co-morbidity, whereas other studies report a higher incidence of seizures in patients with AD. It was previously thought that seizures play a minor role in AD pathophysiology because of their low frequency, and also because they may only be noticed during late stages of AD, suggesting that seizures are likely to be a consequence of neurodegeneration rather than a contributing factor. However, clinical reports indicate that seizures can occur early in the emergence of AD symptoms, particularly in familial AD. In this case, seizures may be an integral part of the emerging pathophysiology. This view has been supported by evidence of recurrent spontaneous seizures in transgenic mouse models of AD in which familial AD is simulated. Additional data from transgenic animals suggest that there may be a much closer relationship between seizures and AD than previously considered. There is also evidence that seizures facilitate production of amyloid β (Aβ) and can cause impairments in cognition and behavior in both animals and humans. However, whether seizures play a role in the early stages of AD pathogenesis is still debated. Therefore, it is timely to review the similarities and differences between AD and epilepsy, as well as data suggesting that seizures may contribute to cognitive and behavioral dysfunction in AD. Here we focus on AD and temporal lobe epilepsy (TLE), a particular type of epilepsy that involves the temporal lobe, a region that influences behavior and is critical to memory. We also consider potential neurobiological mechanisms that support the view that the causes of seizures in TLE may be related to the causes of cognitive dysfunction in AD. We suggest that similar underlying mechanisms may exist for at least some of the aspects of AD that are also found in TLE.
Collapse
Affiliation(s)
- Jeannie Chin
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Helen E. Scharfman
- Child & Adolescent Psychiatry, Physiology & Neuroscience, Psychiatry, New York University Langone Medical Center, New York, NY, USA,Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA,Correspondence to: H.E. Scharfman, Child & Adolescent Psychiatry, Physiology & Neuroscience, Psychiatry, New York University Langone Medical Center, New York, NY, USA. (H.E. Scharfman)
| |
Collapse
|
36
|
Functional and structural properties of dentate granule cells with hilar basal dendrites in mouse entorhino-hippocampal slice cultures. PLoS One 2012; 7:e48500. [PMID: 23144894 PMCID: PMC3492458 DOI: 10.1371/journal.pone.0048500] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 09/26/2012] [Indexed: 12/12/2022] Open
Abstract
During postnatal development hippocampal dentate granule cells (GCs) often extend dendrites from the basal pole of their cell bodies into the hilar region. These so-called hilar basal dendrites (hBD) usually regress with maturation. However, hBDs may persist in a subset of mature GCs under certain conditions (both physiological and pathological). The functional role of these hBD-GCs remains not well understood. Here, we have studied hBD-GCs in mature (≥18 days in vitro) mouse entorhino-hippocampal slice cultures under control conditions and have compared their basic functional properties (basic intrinsic and synaptic properties) and structural properties (dendritic arborisation and spine densities) to those of neighboring GCs without hBDs in the same set of cultures. Except for the presence of hBDs, we did not detect major differences between the two GC populations. Furthermore, paired recordings of neighboring GCs with and without hBDs did not reveal evidence for a heavy aberrant GC-to-GC connectivity. Taken together, our data suggest that in control cultures the presence of hBDs on GCs is neither sufficient to predict alterations in the basic functional and structural properties of these GCs nor indicative of a heavy GC-to-GC connectivity between neighboring GCs.
Collapse
|
37
|
Niv F, Keiner S, Krishna K, Witte OW, Lie DC, Redecker C. Aberrant neurogenesis after stroke: a retroviral cell labeling study. Stroke 2012; 43:2468-75. [PMID: 22738919 DOI: 10.1161/strokeaha.112.660977] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Adult neurogenesis in the dentate gyrus is a unique form of brain plasticity that is strongly stimulated after stroke. We investigate the morphological properties of new granule cells, which are born and develop after the ischemic insult, and query whether these adult-born neurons properly integrate into the pre-existing hippocampal circuitries. METHODS Two well-established models were used to induce either small cortical infarcts (photothrombosis model) or large territorial infarcts (transient middle cerebral artery occlusion model). New granule cells were labeled 4 days after the initial insult by intrahippocampal injection of a retroviral vector encoding green fluorescent protein and newborn neurons were morphologically analyzed using a semiautomatic Neurolucida system and confocal laser scanning microscopy at 6 weeks. RESULTS Approximately 5% to 10% of newborn granule cells displayed significant morphological abnormalities comprising additional basal dendrites and, after middle cerebral artery occlusion, also ectopic cell position. The extent of morphological abnormalities was higher after large territorial infarcts and seems to depend on the severity of ischemic damage. An increased portion of mushroom spines in aberrant neurons suggests stable synaptic integration. However, poststroke generated granule cells with regular appearance also demonstrate alterations in dendritic complexity and spine morphology. CONCLUSIONS The remarkable stimulation of dentate neurogenesis after stroke coincides with an increased rate of aberrantly integrated neurons, which may contribute to functional impairments and, hypothetically, favor pathogenesis of adjustment disorders, cognitive deficits, or epilepsy often seen in stroke patients.
Collapse
Affiliation(s)
- Fanny Niv
- Hans Berger Department of Neurology, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Ma L, Cui XL, Wang Y, Li XW, Yang F, Wei D, Jiang W. Aspirin attenuates spontaneous recurrent seizures and inhibits hippocampal neuronal loss, mossy fiber sprouting and aberrant neurogenesis following pilocarpine-induced status epilepticus in rats. Brain Res 2012; 1469:103-13. [PMID: 22765917 DOI: 10.1016/j.brainres.2012.05.058] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/01/2012] [Accepted: 05/31/2012] [Indexed: 10/28/2022]
Abstract
Accumulating data suggest that inflammation may contribute to epileptogenesis in experimental models as well as in humans. However, whether anti-inflammatory treatments can prevent epileptogenesis still remains controversial. Here, we examined the anti-epileptogenic effect and possible mechanisms of aspirin, a non-selective Cyclooxygenase (COX) inhibitor, in a rat model of lithium-pilocarpine-induced status epilepticus (SE). Epileptic rats were treated with aspirin (20mg/kg) at 0h, 3h, or 24h after the termination of SE, followed by once daily treatment for the subsequent 20 days. We found that aspirin treatment significantly reduced the frequency and duration of spontaneous recurrent seizures during the chronic epileptic phase. Hippocampal neuronal loss five weeks after SE was also attenuated in the CA1, CA3 and hilus following aspirin administration. Furthermore, the aberrant migration of newly generated granule cells and the formation of hilar basal dendrites were prevented by aspirin. Treatment with aspirin starting at 3h or 24h after SE also suppressed the development of mossy fiber sprouting. These findings suggest the possibility of a relative broad time-window for aspirin intervention in the epileptogenic process after injury. Aspirin may serve as a potential adjunctive therapy for individuals susceptible to chronic epilepsy.
Collapse
Affiliation(s)
- Lei Ma
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 17 Changle West Road, Xi'an 710032, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Borschensky CM, Woolley JS, Kipar A, Herden C. Neurogenesis in a Young Dog With Epileptic Seizures. Vet Pathol 2011; 49:766-70. [DOI: 10.1177/0300985811429308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Epileptic seizures can lead to various reactions in the brain, ranging from neuronal necrosis and glial cell activation to focal structural disorganization. Furthermore, increased hippocampal neurogenesis has been documented in rodent models of acute convulsions. This is a report of hippocampal neurogenesis in a dog with spontaneous epileptic seizures. A 16-week-old epileptic German Shepherd Dog had marked neuronal cell proliferation (up to 5 mitotic figures per high-power field and increased immunohistochemical expression of proliferative cell nuclear antigen) in the dentate gyrus accompanied by microglial and astroglial activation. Some granule cells expressed doublecortin, a marker of immature neurons; mitotically active cells expressed neuronal nuclear antigen. No mitotic figures were found in the brain of age-matched control dogs. Whether increased neurogenesis represents a general reaction pattern of young epileptic dogs should be investigated.
Collapse
Affiliation(s)
| | | | - A. Kipar
- Veterinary Pathology, School of Veterinary Science, University of Liverpool, UK
| | - C. Herden
- Institut für Veterinär-Pathologie, Universität Gießen, Germany
| |
Collapse
|
40
|
Seeger N, Zellinger C, Rode A, Roloff F, Bicker G, Russmann V, Fischborn S, Wendt H, Potschka H. The erythropoietin-derived peptide mimetic pHBSP affects cellular and cognitive consequences in a rat post-status epilepticus model. Epilepsia 2011; 52:2333-43. [PMID: 22050420 DOI: 10.1111/j.1528-1167.2011.03302.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The selection of a minimal active sequence of erythropoietin allowed the design of peptide mimetics that exert beneficial effects in the central nervous system but lack an erythropoietic effect. Erythropoietin has been suggested as a promising therapeutic and prophylactic for epilepsies based on its neuroprotective, neuroregenerative, and antiinflammatory potency. Therefore, it is of particular interest to evaluate whether the nonerythropoietic erythropoietin-derived peptide pHBSP can affect epileptogenesis. METHODS In a post-status epilepticus model in rats, we determined the effects of pHBSP and of recombinant human erythropoietin with short-term administration following status epilepticus. KEY FINDINGS Both pHBSP and erythropoietin further enhanced the status epilepticus-associated increase in hippocampal cell proliferation. Thereby, pHBSP seemed to promote neuronal differentiation and survival resulting in a significant increase in neurogenesis. Neither pHBSP nor erythropoietin affected the number of animals exhibiting spontaneous recurrent seizures as well as the seizure frequency in the chronic phase. In the Morris water maze, pHBSP attenuated cognitive deficits in epileptic animals. SIGNIFICANCE In conclusion, the helix B-derived erythropoietin peptide pHBSP can modulate the cellular and cognitive consequences of a status epilepticus. The impact of pHBSP on spatial learning might indicate that the peptide allows beneficial effects on epileptogenesis-associated cognitive deficits. However, it needs to be considered that learning deficits were not abolished by pHBSP and that the effects were not observed consistently until the end of the study. Therefore, adjustment of timing, duration, and dose of peptide administration might be necessary to further evaluate the efficacy of pHBSP.
Collapse
Affiliation(s)
- Natalie Seeger
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Impact of the erythropoietin-derived peptide mimetic Epotris on the histopathological consequences of status epilepticus. Epilepsy Res 2011; 96:241-9. [DOI: 10.1016/j.eplepsyres.2011.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/09/2011] [Accepted: 06/11/2011] [Indexed: 11/18/2022]
|
42
|
Impact of the NCAM derived mimetic peptide plannexin on the acute cellular consequences of a status epilepticus. Neurosci Lett 2011; 501:173-8. [PMID: 21787839 DOI: 10.1016/j.neulet.2011.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/27/2011] [Accepted: 07/06/2011] [Indexed: 11/22/2022]
Abstract
Plannexin represents a NCAM-derived peptide mimicking trans-homophilic NCAM interaction, which proved to exert neuroprotective effects in vitro. The effect of plannexin was evaluated in a rat status epilepticus model. As expected, prolonged seizure activity resulted in a pronounced cell loss in hippocampal subregions. The comparison between the vehicle- and plannexin-treated animals with status epilepticus did not reveal neuroprotective effects of plannexin on mature neurons. However, treatment with plannexin partially prevented the reduction in the number of doublecortin-labeled neuronal progenitor cells, which was evident 48h following status epilepticus. In conclusion, the data might give first evidence that plannexin can protect immature neurons in vivo. Future studies are necessary to evaluate whether disease-modifying or preventive effects are observed in models of epileptogenesis.
Collapse
|
43
|
Aasebø IEJ, Blankvoort S, Tashiro A. Critical maturational period of new neurons in adult dentate gyrus for their involvement in memory formation. Eur J Neurosci 2011; 33:1094-100. [PMID: 21395853 DOI: 10.1111/j.1460-9568.2011.07608.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Adult dentate gyrus produces new neurons continuously throughout life. Multiple lines of evidence have pointed to the possibility that young neurons during a certain maturational stage mediate an important role in memory processing. In this review, we highlight the existing evidence of a 'critical period' for new neurons in their involvement in memory formation, describe the unique properties of young neurons as potential mechanisms underlying the critical period, and discuss the implications of the critical period for the function of adult neurogenesis.
Collapse
Affiliation(s)
- Ida E J Aasebø
- Kavli Institute for Systems Neuroscience and Centre for the Biology of Memory, Faculty of Medicine, Norwegian University of Science and Technology and St. Olavs Hospital, 7030 Trondheim, Norway
| | | | | |
Collapse
|
44
|
Pierce JP, McCloskey DP, Scharfman HE. Morphometry of hilar ectopic granule cells in the rat. J Comp Neurol 2011; 519:1196-218. [PMID: 21344409 DOI: 10.1002/cne.22568] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Granule cell (GC) neurogenesis in the dentate gyrus (DG) does not always proceed normally. After severe seizures (e.g., status epilepticus [SE]) and some other conditions, newborn GCs appear in the hilus. Hilar ectopic GCs (EGCs) can potentially provide insight into the effects of abnormal location and seizures on GC development. Additionally, hilar EGCs that develop after SE may contribute to epileptogenesis and cognitive impairments that follow SE. Thus, it is critical to understand how EGCs differ from normal GCs. Relatively little morphometric information is available on EGCs, especially those restricted to the hilus. This study quantitatively analyzed the structural morphology of hilar EGCs from adult male rats several months after pilocarpine-induced SE, when they are considered to have chronic epilepsy. Hilar EGCs were physiologically identified in slices, intracellularly labeled, processed for light microscopic reconstruction, and compared to GC layer GCs, from both the same post-SE tissue and the NeuroMorpho database (normal GCs). Consistently, hilar EGC and GC layer GCs had similar dendritic lengths and field sizes, and identifiable apical dendrites. However, hilar EGC dendrites were topologically more complex, with more branch points and tortuous dendritic paths. Three-dimensional analysis revealed that, remarkably, hilar EGC dendrites often extended along the longitudinal DG axis, suggesting increased capacity for septotemporal integration. Axonal reconstruction demonstrated that hilar EGCs contributed to mossy fiber sprouting. This combination of preserved and aberrant morphological features, potentially supporting convergent afferent input to EGCs and broad, divergent efferent output, could help explain why the hilar EGC population could impair DG function.
Collapse
Affiliation(s)
- Joseph P Pierce
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York 10065, USA.
| | | | | |
Collapse
|
45
|
Pugh P, Adlaf E, Zhao CS, Markwardt S, Gavin C, Wadiche J, Overstreet-Wadiche L. Enhanced integration of newborn neurons after neonatal insults. Front Neurosci 2011; 5:45. [PMID: 21490706 PMCID: PMC3070953 DOI: 10.3389/fnins.2011.00045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 03/17/2011] [Indexed: 01/16/2023] Open
Abstract
The production and integration of adult-generated neurons in the dentate gyrus is dramatically perturbed by a variety of pathological insults, including repetitive seizures and hypoxia/ischemia. Less is known about how insults affect early postnatal neurogenesis, during the developmental period when the majority of dentate neurons are produced. Here we tested how single episodes of hypoxia or chemically induced seizure activity in postnatal day 10 mice alter granule cell production and integration. Although neither insult was sufficient to alter the number of newborn neurons nor the population of proliferating cells, both treatments increased the dendritic complexity of newborn granule cells that were born around the time of the insult. Surprisingly, only the dendritic enhancement caused by hypoxia was associated with increased synaptic integration. These results suggest that alterations in dendritic integration can be dissociated from altered neural production and that integration appears to have a lower threshold for perturbation. Furthermore, newborn neurons in adult mice that experienced neonatal hypoxia had reduced dendritic length while having no alterations in number. Together these results suggest that single insults during the neonatal period can have both long- and short-term consequences for neuronal maturation.
Collapse
Affiliation(s)
- Phyllis Pugh
- McKnight Brain Institute and Department of Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Time-lapse imaging of disease progression in deep brain areas using fluorescence microendoscopy. Nat Med 2011; 17:223-8. [PMID: 21240263 PMCID: PMC3833825 DOI: 10.1038/nm.2292] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 10/07/2010] [Indexed: 12/11/2022]
Abstract
The combination of intravital microscopy and animal models of disease has propelled studies of disease mechanisms and treatments. However, many disorders afflict tissues inaccessible to light microscopy in live subjects. Here we introduce cellular-level time-lapse imaging deep within the live mammalian brain by one- and two-photon fluorescence microendoscopy over multiple weeks. Bilateral imaging sites allowed longitudinal comparisons within individual subjects, including of normal and diseased tissues. Using this approach, we tracked CA1 hippocampal pyramidal neuron dendrites in adult mice, revealing these dendrites' extreme stability and rare examples of their structural alterations. To illustrate disease studies, we tracked deep lying gliomas by observing tumor growth, visualizing three-dimensional vasculature structure and determining microcirculatory speeds. Average erythrocyte speeds in gliomas declined markedly as the disease advanced, notwithstanding significant increases in capillary diameters. Time-lapse microendoscopy will be applicable to studies of numerous disorders, including neurovascular, neurological, cancerous and trauma-induced conditions.
Collapse
|
47
|
Murphy BL, Pun RYK, Yin H, Faulkner CR, Loepke AW, Danzer SC. Heterogeneous integration of adult-generated granule cells into the epileptic brain. J Neurosci 2011; 31:105-17. [PMID: 21209195 PMCID: PMC3022369 DOI: 10.1523/jneurosci.2728-10.2011] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 09/08/2010] [Accepted: 10/21/2010] [Indexed: 11/21/2022] Open
Abstract
The functional impact of adult-generated granule cells in the epileptic brain is unclear, with data supporting both protective and maladaptive roles. These conflicting findings could be explained if new granule cells integrate heterogeneously, with some cells taking neutral or adaptive roles and others contributing to recurrent circuitry supporting seizures. Here, we tested this hypothesis by completing detailed morphological characterizations of age- and experience-defined cohorts of adult-generated granule cells from transgenic mice. The majority of newborn cells exposed to an epileptogenic insult exhibited reductions in dendritic spine number, suggesting reduced excitatory input to these cells. A significant subset, however, exhibited higher spine numbers. These latter cells tended to have enlarged cell bodies, long basal dendrites, or both. Moreover, cells with basal dendrites received significantly more recurrent mossy fiber input through their apical dendrites, indicating that these cells are robustly integrated into the pathological circuitry of the epileptic brain. These data imply that newborn cells play complex--and potentially conflicting--roles in epilepsy.
Collapse
Affiliation(s)
- Brian L. Murphy
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Program in Neuroscience, University of Cincinnati, Cincinnati, Ohio 45229
| | - Raymund Y. K. Pun
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Hulian Yin
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Christian R. Faulkner
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Andreas W. Loepke
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, Ohio 45221, and
- Program in Neuroscience, University of Cincinnati, Cincinnati, Ohio 45229
| | - Steve C. Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, Ohio 45221, and
- Program in Neuroscience, University of Cincinnati, Cincinnati, Ohio 45229
| |
Collapse
|
48
|
Kress GJ, Dowling MJ, Eisenman LN, Mennerick S. Axonal sodium channel distribution shapes the depolarized action potential threshold of dentate granule neurons. Hippocampus 2010; 20:558-71. [PMID: 19603521 DOI: 10.1002/hipo.20667] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intrinsic excitability is a key feature dictating neuronal response to synaptic input. Here we investigate the recent observation that dentate granule neurons exhibit a more depolarized voltage threshold for action potential initiation than CA3 pyramidal neurons. We find no evidence that tonic GABA currents, leak or voltage-gated potassium conductances, or the expression of sodium channel isoform differences can explain this depolarized threshold. Axonal initial segment voltage-gated sodium channels, which are dominated by the Na(V)1.6 isoform in both cell types, distribute more proximally and exhibit lower overall density in granule neurons than in CA3 neurons. To test possible contributions of sodium channel distributions to voltage threshold and to test whether morphological differences participate, we performed simulations of dentate granule neurons and of CA3 pyramidal neurons. These simulations revealed that cell morphology and sodium channel distribution combine to yield the characteristic granule neuron action potential upswing and voltage threshold. Proximal axon sodium channel distribution strongly contributes to the higher voltage threshold of dentate granule neurons for two reasons. First, action potential initiation closer to the somatodendritic current sink causes the threshold of the initiating axon compartment to rise. Second, the proximity of the action potential initiation site to the recording site causes somatic recordings to more faithfully reflect the depolarized threshold of the axon than in cells like CA3 neurons, with distally initiating action potentials. Our results suggest that the proximal location of axon sodium channels in dentate granule neurons contributes to the intrinsic excitability differences between DG and CA3 neurons and may participate in the low-pass filtering function of dentate granule neurons.
Collapse
Affiliation(s)
- Geraldine J Kress
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | |
Collapse
|
49
|
Pekcec A, Lüpke M, Baumann R, Seifert H, Potschka H. Modulation of neurogenesis by targeted hippocampal irradiation fails to affect kindling progression. Hippocampus 2010; 21:866-76. [PMID: 20865736 DOI: 10.1002/hipo.20802] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2010] [Indexed: 12/31/2022]
Abstract
Changes in the rate of dentate granule cell neurogenesis and in the fate of newborn granule cells have been implicated in the development and progression of epilepsies. Strategies to normalize neurogenesis in chronic epilepsy models are thought to increase our understanding of the functional consequences of aberrant neurogenesis in the epileptic brain. Therefore, we modulated neurogenesis in an amygdala kindling paradigm in rats by targeted irradiation of the hippocampus using a medical linear accelerator device. Selective irradiation normalized the hippocampal cell proliferation rate in kindled animals. Both, in kindled and nonkindled rats the number of BrdU/NeuN-labeled newborn neurons was reduced in response to irradiation. Whereas kindling resulted in a pronounced increase in the number of neuroblasts identified based on doublecortin-labeling, irradiation prevented the expansion of the neuroblast population. Moreover, irradiation counteracted the kindling-associated increase in hilar basal dendrites, and kept the fraction of cells with basal dendrites at control levels. Despite the efficacious modulation of neurogenesis, irradiation did not affect the rate of kindling progression. Both, the number of stimulations as well as the cumulative afterdischarge duration to reach respective seizure stages were comparable in animals with and without irradiation. In addition, pre- and postkindling thresholds as well as seizure parameters recorded at threshold stimulation remained unaffected by irradiation. In conclusion, the fact that the efficacious modulation of neurogenesis by irradiation did not exert any effects on kindling acquisition and kindled seizures suggests that newborn neurons do not critically contribute to the hyperexcitable state in the chronic epilepsy model used.
Collapse
Affiliation(s)
- Anton Pekcec
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | |
Collapse
|
50
|
Herculano-Houzel S. Coordinated scaling of cortical and cerebellar numbers of neurons. Front Neuroanat 2010; 4:12. [PMID: 20300467 PMCID: PMC2839851 DOI: 10.3389/fnana.2010.00012] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 02/18/2010] [Indexed: 11/13/2022] Open
Abstract
While larger brains possess concertedly larger cerebral cortices and cerebella, the relative size of the cerebral cortex increases with brain size, but relative cerebellar size does not. In the absence of data on numbers of neurons in these structures, this discrepancy has been used to dispute the hypothesis that the cerebral cortex and cerebellum function and have evolved in concert and to support a trend towards neocorticalization in evolution. However, the rationale for interpreting changes in absolute and relative size of the cerebral cortex and cerebellum relies on the assumption that they reflect absolute and relative numbers of neurons in these structures across all species - an assumption that our recent studies have shown to be flawed. Here I show for the first time that the numbers of neurons in the cerebral cortex and cerebellum are directly correlated across 19 mammalian species of four different orders, including humans, and increase concertedly in a similar fashion both within and across the orders Eulipotyphla (Insectivora), Rodentia, Scandentia and Primata, such that on average a ratio of 3.6 neurons in the cerebellum to every neuron in the cerebral cortex is maintained across species. This coordinated scaling of cortical and cerebellar numbers of neurons provides direct evidence in favor of concerted function, scaling and evolution of these brain structures, and suggests that the common notion that equates cognitive advancement with neocortical expansion should be revisited to consider in its stead the coordinated scaling of neocortex and cerebellum as a functional ensemble.
Collapse
Affiliation(s)
- Suzana Herculano-Houzel
- Laboratório de Neuroanatomia Comparada, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro-RJ, Brazil
| |
Collapse
|