1
|
Drake AW, Jerow LG, Ruksenas JV, McCoy C, Danzer SC. Somatostatin interneuron fate-mapping and structure in a Pten knockout model of epilepsy. Front Cell Neurosci 2024; 18:1474613. [PMID: 39497922 PMCID: PMC11532043 DOI: 10.3389/fncel.2024.1474613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Disruption of inhibitory interneurons is common in the epileptic brain and is hypothesized to play a pivotal role in epileptogenesis. Abrupt disruption and loss of interneurons is well-characterized in status epilepticus models of epilepsy, however, status epilepticus is a relatively rare cause of epilepsy in humans. How interneuron disruption evolves in other forms of epilepsy is less clear. Here, we explored how somatostatin (SST) interneuron disruption evolves in quadruple transgenic Gli1-CreERT2, Ptenfl/fl, SST-FlpO, and frt-eGFP mice. In these animals, epilepsy develops following deletion of the mammalian target of rapamycin (mTOR) negative regulator phosphatase and tensin homolog (Pten) from a subset of dentate granule cells, while downstream Pten-expressing SST neurons are fate-mapped with green fluorescent protein (GFP). The model captures the genetic complexity of human mTORopathies, in which mutations can be restricted to excitatory neuron lineages, implying that interneuron involvement is later developing and secondary. In dentate granule cell (DGC)-Pten knockouts (KOs), the density of fate-mapped SST neurons was reduced in the hippocampus, but their molecular phenotype was unchanged, with similar percentages of GFP+ cells immunoreactive for SST and parvalbumin (PV). Surviving SST neurons in the dentate gyrus had larger somas, and the density of GFP+ processes in the dentate molecular layer was unchanged despite SST cell loss and expansion of the molecular layer, implying compensatory sprouting of surviving cells. The density of Znt3-immunolabeled puncta, a marker of granule cell presynaptic terminals, apposed to GFP+ processes in the hilus was increased, suggesting enhanced granule cell input to SST neurons. Finally, the percentage of GFP+ cells that were FosB positive was significantly increased, implying that surviving SST neurons are more active. Together, findings suggest that somatostatin-expressing interneurons exhibit a combination of pathological (cell loss) and adaptive (growth) responses to hyperexcitability and seizures driven by upstream Pten KO excitatory granule cells.
Collapse
Affiliation(s)
- Austin W. Drake
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lilian G. Jerow
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Justin V. Ruksenas
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Carlie McCoy
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Steve C. Danzer
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Neuroscience Research Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
2
|
Guo D, Han L, Godale CM, Rensing NR, Danzer SC, Wong M. A role of dentate gyrus mechanistic target of rapamycin activation in epileptogenesis in a mouse model of posttraumatic epilepsy. Epilepsia 2024; 65:2127-2137. [PMID: 38761065 PMCID: PMC11251851 DOI: 10.1111/epi.18011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/20/2024]
Abstract
OBJECTIVE The mechanistic target of rapamycin (mTOR) pathway has been implicated in promoting epileptogenesis in animal models of acquired epilepsy, such as posttraumatic epilepsy (PTE) following traumatic brain injury (TBI). However, the specific anatomical regions and neuronal populations mediating mTOR's role in epileptogenesis are not well defined. In this study, we tested the hypothesis that mTOR activation in dentate gyrus granule cells promotes neuronal death, mossy fiber sprouting, and PTE in the controlled cortical impact (CCI) model of TBI. METHODS An adeno-associated virus (AAV)-Cre viral vector was injected into the hippocampus of Rptorflox/flox (regulatory-associated protein of mTOR) mutant mice to inhibit mTOR activation in dentate gyrus granule cells. Four weeks after AAV-Cre or AAV-vehicle injection, mice underwent CCI injury and were subsequently assessed for mTOR pathway activation by Western blotting, neuronal death, and mossy fiber sprouting by immunopathological analysis, and posttraumatic seizures by video-electroencephalographic monitoring. RESULTS AAV-Cre injection primarily affected the dentate gyrus and inhibited hippocampal mTOR activation following CCI injury. AAV-Cre-injected mice had reduced neuronal death in dentate gyrus detected by Fluoro-Jade B staining and decreased mossy fiber sprouting by ZnT3 immunostaining. Finally, AAV-Cre-injected mice exhibited a decrease in incidence of PTE. SIGNIFICANCE mTOR pathway activation in dentate gyrus granule cells may at least partly mediate pathological abnormalities and epileptogenesis in models of TBI and PTE. Targeted modulation of mTOR activity in this hippocampal network may represent a focused therapeutic approach for antiepileptogenesis and prevention of PTE.
Collapse
Affiliation(s)
- Dongjun Guo
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Lirong Han
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Christin M. Godale
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Nicholas R. Rensing
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Steve C. Danzer
- Department of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Michael Wong
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA 63110
| |
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
|
McGann AM, Westerkamp GC, Chalasani A, Danzer CSK, Parkins EV, Rajathi V, Horn PS, Pedapati EV, Tiwari D, Danzer SC, Gross C. MiR-324-5p inhibition after intrahippocampal kainic acid-induced status epilepticus does not prevent epileptogenesis in mice. Front Neurol 2023; 14:1280606. [PMID: 38033777 PMCID: PMC10687438 DOI: 10.3389/fneur.2023.1280606] [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: 08/20/2023] [Accepted: 10/20/2023] [Indexed: 12/02/2023] Open
Abstract
Background Acquired epilepsies are caused by an initial brain insult that is followed by epileptogenesis and finally the development of spontaneous recurrent seizures. The mechanisms underlying epileptogenesis are not fully understood. MicroRNAs regulate mRNA translation and stability and are frequently implicated in epilepsy. For example, antagonism of a specific microRNA, miR-324-5p, before brain insult and in a model of chronic epilepsy decreases seizure susceptibility and frequency, respectively. Here, we tested whether antagonism of miR-324-5p during epileptogenesis inhibits the development of epilepsy. Methods We used the intrahippocampal kainic acid (IHpKa) model to initiate epileptogenesis in male wild type C57BL/6 J mice aged 6-8 weeks. Twenty-four hours after IHpKa, we administered a miR-324-5p or scrambled control antagomir intracerebroventricularly and implanted cortical surface electrodes for EEG monitoring. EEG data was collected for 28 days and analyzed for seizure frequency and duration, interictal spike activity, and EEG power. Brains were collected for histological analysis. Results Histological analysis of brain tissue showed that IHpKa caused characteristic hippocampal damage in most mice regardless of treatment. Antagomir treatment did not affect latency to, frequency, or duration of spontaneous recurrent seizures or interictal spike activity but did alter the temporal development of frequency band-specific EEG power. Conclusion These results suggest that miR-324-5p inhibition during epileptogenesis induced by status epilepticus does not convey anti-epileptogenic effects despite having subtle effects on EEG frequency bands. Our results highlight the importance of timing of intervention across epilepsy development and suggest that miR-324-5p may act primarily as a proconvulsant rather than a pro-epileptogenic regulator.
Collapse
Affiliation(s)
- Amanda M. McGann
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Grace C. Westerkamp
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Alisha Chalasani
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Cole S. K. Danzer
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Emma V. Parkins
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Valerine Rajathi
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Paul S. Horn
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Ernest V. Pedapati
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Steve C. Danzer
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Anesthesia, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Anesthesia, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christina Gross
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
5
|
Tipton AE, Del Angel YC, Hixson K, Carlsen J, Strode D, Busquet N, Mesches MH, Gonzalez MI, Napoli E, Russek SJ, Brooks-Kayal AR. Selective Neuronal Knockout of STAT3 Function Inhibits Epilepsy Progression, Improves Cognition, and Restores Dysregulated Gene Networks in a Temporal Lobe Epilepsy Model. Ann Neurol 2023; 94:106-122. [PMID: 36935347 PMCID: PMC10313781 DOI: 10.1002/ana.26644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
OBJECTIVE Temporal lobe epilepsy (TLE) is a progressive disorder mediated by pathological changes in molecular cascades and hippocampal neural circuit remodeling that results in spontaneous seizures and cognitive dysfunction. Targeting these cascades may provide disease-modifying treatments for TLE patients. Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) inhibitors have emerged as potential disease-modifying therapies; a more detailed understanding of JAK/STAT participation in epileptogenic responses is required, however, to increase the therapeutic efficacy and reduce adverse effects associated with global inhibition. METHODS We developed a mouse line in which tamoxifen treatment conditionally abolishes STAT3 signaling from forebrain excitatory neurons (nSTAT3KO). Seizure frequency (continuous in vivo electroencephalography) and memory (contextual fear conditioning and motor learning) were analyzed in wild-type and nSTAT3KO mice after intrahippocampal kainate (IHKA) injection as a model of TLE. Hippocampal RNA was obtained 24 h after IHKA and subjected to deep sequencing. RESULTS Selective STAT3 knock-out in excitatory neurons reduced seizure progression and hippocampal memory deficits without reducing the extent of cell death or mossy fiber sprouting induced by IHKA injection. Gene expression was rescued in major networks associated with response to brain injury, neuronal plasticity, and learning and memory. We also provide the first evidence that neuronal STAT3 may directly influence brain inflammation. INTERPRETATION Inhibiting neuronal STAT3 signaling improved outcomes in an animal model of TLE, prevented progression of seizures and cognitive co-morbidities while rescuing pathogenic changes in gene expression of major networks associated with epileptogenesis. Specifically targeting neuronal STAT3 may be an effective disease-modifying strategy for TLE. ANN NEUROL 2023;94:106-122.
Collapse
Affiliation(s)
- Allison E. Tipton
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Yasmin Cruz Del Angel
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kathryn Hixson
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jessica Carlsen
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Dana Strode
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicolas Busquet
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael H. Mesches
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marco I. Gonzalez
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Eleonora Napoli
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Shelley J. Russek
- Graduate Program for Neuroscience, Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Amy R. Brooks-Kayal
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
6
|
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
|
7
|
Puhahn-Schmeiser B, Kleemann T, Jabbarli R, Bock HH, Beck J, Freiman TM. Granule cell dispersion in two mouse models of temporal lobe epilepsy and reeler mice is associated with changes in dendritic orientation and spine distribution. Hippocampus 2022; 32:517-528. [PMID: 35621370 DOI: 10.1002/hipo.23447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 11/06/2022]
Abstract
Temporal lobe epilepsy is characterized by hippocampal neuronal death in CA1 and hilus. Dentate gyrus granule cells survive but show dispersion of the compact granule cell layer. This is associated with decrease of the glycoprotein Reelin, which regulates neuron migration and dendrite outgrow. Reelin-deficient (reeler) mice show no layering, their granule cells are dispersed throughout the dentate gyrus. We studied granule cell dendritic orientation and distribution of postsynaptic spines in reeler mice and two mouse models of temporal lobe epilepsy, namely the p35 knockout mice, which show Reelin-independent neuronal migration defects, and mice with unilateral intrahippocampal kainate injection. Granule cells were Golgi-stained and analyzed, using a computerized camera lucida system. Granule cells in naive controls exhibited a vertically oriented dendritic arbor with a small bifurcation angle if positioned proximal to the hilus and a wider dendritic bifurcation angle, if positioned distally. P35 knockout- and kainate-injected mice showed a dispersed granule cell layer, granule cells showed basal dendrites with wider bifurcation angles, which lost position-specific differences. Reeler mice lacked dendritic orientation. P35 knockout- and kainate-injected mice showed increased dendritic spine density in the granule cell layer. Molecular layer dendrites showed a reduced spine density in kainate-injected mice only, whereas in p35 knockouts no reduced spine density was seen. Reeler mice showed a homogenous high spine density. We hypothesize that granule cells migrate in temporal lobe epilepsy, develop new dendrites which show a spread of the dendritic tree, create new spines in areas proximal to mossy fiber sprouting, which is present in p35 knockout- and kainate-injected mice and loose spines on distal dendrites if mossy cell death is present, as it was in kainate-injected mice only. These results are in accordance with findings in epilepsy patients.
Collapse
Affiliation(s)
- Barbara Puhahn-Schmeiser
- Faculty of Medicine, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| | - Tobias Kleemann
- Department of Gastroenterology and Rheumatology, Carl-Thiem-Hospital, Cottbus, Germany
| | - Ramazan Jabbarli
- Faculty of Medicine, Department of Neurosurgery, Medical Center, University of Duisburg-Essen, Essen, Germany
| | - Hans H Bock
- Faculty of Medicine, Department of Gastroenterology, Hepatology and Infectiology, Medical Center, University of Duesseldorf, Duesseldorf, Germany
| | - Jürgen Beck
- Faculty of Medicine, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas M Freiman
- Department of Neurosurgery, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
8
|
Łowicki D, Przybylski P. Tandem construction of biological relevant aliphatic 5-membered N-heterocycles. Eur J Med Chem 2022; 235:114303. [PMID: 35344904 DOI: 10.1016/j.ejmech.2022.114303] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/20/2022]
Abstract
Nature often uses cascade reactions in a highly stereocontrolled manner for assembly structurally diverse nitrogen-containing heterocyclic scaffolds, i.e. secondary metabolites, important for medicinal chemistry and pharmacy. Five-membered nitrogen-containing heterocycles as standalone rings, as well as spiro and polycyclic systems are pharmacophores of drugs approved in various therapies, i.a. antibacterial or antiviral, antifungal, anticancer, antidiabetic, as they target many key enzymes. Furthermore, a large number of pyrrolidine derivatives are currently considered as drug candidates. Cascade transformations, also known as domino or tandem reactions, offer straightforward methods to build N-heterocyclic libraries of the great structural variety desired for drawing SAR conclusions. The tandem transformations are often atom economic and time-saving because they are performed as the one-pot, so no need for purification after each 'virtual' step and the limited necessity of protective groups are characteristic for these processes. Thus, the same results as in classical multistep synthesis can be achieved at markedly lower costs and shorter time, which is in line with modern green chemistry rules. Great advantage of cascade reactions is often reflected in their high regio- and stereoselectivities, enabling the preparing of the heterocyclic compound better fitted to the expected target in cells. This review reveals the biological relevance of N-heterocyclic scaffolds based on saturated 5-membered rings since we showed a number of examples of approved drugs together with the recent biologically attractive leading structures of drug candidates. Next, novel cascade synthetic procedures, taking into account the structure of the reactants and reaction mechanisms, enabling to obtain biological-relevant heterocyclic frameworks with good yields and relatively high stereoselectivity, were reviewed and compared. The review covers the advances of designing biological active N-heterocycles mainly from 2018 to 2021, whereas the synthetic part is focused on the last 7 years.
Collapse
Affiliation(s)
- Daniel Łowicki
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614, Poznan, Poland
| | - Piotr Przybylski
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614, Poznan, Poland.
| |
Collapse
|
9
|
Chen P, Chen F, Wu Y, Zhou B. New Insights Into the Role of Aberrant Hippocampal Neurogenesis in Epilepsy. Front Neurol 2022; 12:727065. [PMID: 34975709 PMCID: PMC8714646 DOI: 10.3389/fneur.2021.727065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
Data accumulated over the past four decades have confirmed that adult hippocampal neurogenesis (HN) plays a key role in the wide spectrum of hippocampal pathology. Epilepsy is a disorder of the central nervous system characterized by spontaneous recurrent seizures. Although neurogenesis in persistent germinative zones is altered in the adult rodent models of epilepsy, the effects of seizure-induced neurogenesis in the epileptic brain, in terms of either a pathological or reparative role, are only beginning to be explored. In this review, we described the most recent advances in neurogenesis in epilepsy and outlooked future directions for neural stem cells (NSCs) and epilepsy-in-a-dish models. We proposed that it may help in refining the underlying molecular mechanisms of epilepsy and improving the therapies and precision medicine for patients with epilepsy.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fuchao Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Yue Wu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Penning A, Tosoni G, Abiega O, Bielefeld P, Gasperini C, De Pietri Tonelli D, Fitzsimons CP, Salta E. Adult Neural Stem Cell Regulation by Small Non-coding RNAs: Physiological Significance and Pathological Implications. Front Cell Neurosci 2022; 15:781434. [PMID: 35058752 PMCID: PMC8764185 DOI: 10.3389/fncel.2021.781434] [Citation(s) in RCA: 7] [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: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
The adult neurogenic niches are complex multicellular systems, receiving regulatory input from a multitude of intracellular, juxtacrine, and paracrine signals and biological pathways. Within the niches, adult neural stem cells (aNSCs) generate astrocytic and neuronal progeny, with the latter predominating in physiological conditions. The new neurons generated from this neurogenic process are functionally linked to memory, cognition, and mood regulation, while much less is known about the functional contribution of aNSC-derived newborn astrocytes and adult-born oligodendrocytes. Accumulating evidence suggests that the deregulation of aNSCs and their progeny can impact, or can be impacted by, aging and several brain pathologies, including neurodevelopmental and mood disorders, neurodegenerative diseases, and also by insults, such as epileptic seizures, stroke, or traumatic brain injury. Hence, understanding the regulatory underpinnings of aNSC activation, differentiation, and fate commitment could help identify novel therapeutic avenues for a series of pathological conditions. Over the last two decades, small non-coding RNAs (sncRNAs) have emerged as key regulators of NSC fate determination in the adult neurogenic niches. In this review, we synthesize prior knowledge on how sncRNAs, such as microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), may impact NSC fate determination in the adult brain and we critically assess the functional significance of these events. We discuss the concepts that emerge from these examples and how they could be used to provide a framework for considering aNSC (de)regulation in the pathogenesis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Caterina Gasperini
- Neurobiology of miRNAs Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Carlos P. Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
11
|
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
|
12
|
Wulsin AC, Kraus KL, Gaitonde KD, Suru V, Arafa SR, Packard BA, Herman JP, Danzer SC. The glucocorticoid receptor specific modulator CORT108297 reduces brain pathology following status epilepticus. Exp Neurol 2021; 341:113703. [PMID: 33745919 PMCID: PMC8169587 DOI: 10.1016/j.expneurol.2021.113703] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Glucocorticoid levels rise rapidly following status epilepticus and remain elevated for weeks after the injury. To determine whether glucocorticoid receptor activation contributes to the pathological sequelae of status epilepticus, mice were treated with a novel glucocorticoid receptor modulator, C108297. METHODS Mice were treated with either C108297 or vehicle for 10 days beginning one day after pilocarpine-induced status epilepticus. Baseline and stress-induced glucocorticoid secretion were assessed to determine whether hypothalamic-pituitary-adrenal axis hyperreactivity could be controlled. Status epilepticus-induced pathology was assessed by quantifying ectopic hippocampal granule cell density, microglial density, astrocyte density and mossy cell loss. Neuronal network function was examined indirectly by determining the density of Fos immunoreactive neurons following restraint stress. RESULTS Treatment with C108297 attenuated corticosterone hypersecretion after status epilepticus. Treatment also decreased the density of hilar ectopic granule cells and reduced microglial proliferation. Mossy cell loss, on the other hand, was not prevented in treated mice. C108297 altered the cellular distribution of Fos protein but did not restore the normal pattern of expression. INTERPRETATION Results demonstrate that baseline corticosterone levels can be normalized with C108297, and implicate glucocorticoid signaling in the development of structural changes following status epilepticus. These findings support the further development of glucocorticoid receptor modulators as novel therapeutics for the prevention of brain pathology following status epilepticus.
Collapse
Affiliation(s)
- Aynara C Wulsin
- Cincinnati Children's Hospital Medical Center, Department of Anesthesia, USA; Cincinnati Children's Hospital Medical Center, Department of Pediatrics, USA; University of Cincinnati, Medical Scientist Training Program, USA; University of Cincinnati, Neuroscience Graduate Program, USA
| | - Kimberly L Kraus
- Cincinnati Children's Hospital Medical Center, Department of Anesthesia, USA; University of Cincinnati, Medical Scientist Training Program, USA; University of Cincinnati, Neuroscience Graduate Program, USA
| | - Kevin D Gaitonde
- University of Cincinnati, Medical Scientist Training Program, USA
| | - Venkat Suru
- Cincinnati Children's Hospital Medical Center, Department of Anesthesia, USA
| | - Salwa R Arafa
- Cincinnati Children's Hospital Medical Center, Department of Anesthesia, USA
| | - Benjamin A Packard
- University of Cincinnati, Department of Pharmacology & Systems Physiology
| | - James P Herman
- University of Cincinnati, Department of Pharmacology & Systems Physiology
| | - Steve C Danzer
- Cincinnati Children's Hospital Medical Center, Department of Anesthesia, USA; Cincinnati Children's Hospital Medical Center, Department of Pediatrics, USA; University of Cincinnati, Medical Scientist Training Program, USA; University of Cincinnati, Neuroscience Graduate Program, USA.
| |
Collapse
|
13
|
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
|
14
|
FGFR Regulation of Dendrite Elaboration in Adult-born Granule Cells Depends on Intracellular Mediator and Proximity to the Soma. Neuroscience 2020; 453:148-167. [PMID: 33246055 DOI: 10.1016/j.neuroscience.2020.10.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/24/2023]
Abstract
Fibroblast Growth Factor Receptors (FGFRs) play crucial roles in promoting dendrite growth and branching during development. In mice, three FGFR genes, Fgfr1, Fgfr2, and Fgfr3, remain expressed in the adult neurogenic niche of the hippocampal dentate gyrus. However, the function of FGFRs in the dendritic maturation of adult-born neurons remains largely unexplored. Here, using conditional alleles of Fgfr1, Fgfr2, and Fgfr3 as well as Fgfr1 alleles lacking binding sites for Phospholipase-Cγ (PLCγ) and FGF Receptor Substrate (FRS) proteins, we test the requirement for FGFRs in dendritogenesis of adult-born granule cells. We find that deleting all three receptors results in a small decrease in proximal dendrite elaboration. In contrast, specifically mutating Tyr766 in FGFR1 (a PLCγ binding site) in an Fgfr2;Fgfr3 deficient background results in a dramatic increase of overall dendrite elaboration, while blocking FGFR1-FRS signaling causes proximal dendrite deficits and, to a lesser extent than Tyr766 mutants, increases distal dendrite elaboration. These findings reveal unexpectedly complex roles for FGFRs and their intracellular mediators in regulating proximal and distal dendrite elaboration, with the most notable role in suppressing distal elaboration through the PLCγbinding site.
Collapse
|
15
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
16
|
|
17
|
Li J, Leverton LK, Naganatanahalli LM, Christian-Hinman CA. Seizure burden fluctuates with the female reproductive cycle in a mouse model of chronic temporal lobe epilepsy. Exp Neurol 2020; 334:113492. [PMID: 33007292 DOI: 10.1016/j.expneurol.2020.113492] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022]
Abstract
Women with catamenial epilepsy often experience increased seizure burden near the time of ovulation (periovulatory) or menstruation (perimenstrual). To date, a rodent model of chronic temporal lobe epilepsy (TLE) that exhibits similar endogenous fluctuations in seizures has not been identified. Here, we investigated whether seizure burden changes with the estrous cycle in the intrahippocampal kainic acid (IHKA) mouse model of TLE. Adult female IHKA mice and saline-injected controls were implanted with EEG electrodes in the ipsilateral hippocampus. At one and two months post-injection, 24/7 video-EEG recordings were collected and estrous cycle stage was assessed daily. Seizures were detected using a custom convolutional neural network machine learning process. Seizure burden was compared within each mouse between diestrus and combined proestrus and estrus days (pro/estrus) at two months post-injection. IHKA mice showed higher seizure burden on pro/estrus compared with diestrus, characterized by increased time in seizures and longer seizure duration. When all IHKA mice were included, no group differences were observed in seizure frequency or EEG power. However, increased baseline seizure burden on diestrus was correlated with larger cycle-associated differences, and when analyses were restricted to mice that showed the severe epilepsy typical of the IHKA model, increased seizure frequency on pro/estrus was also revealed. Controls showed no differences in EEG parameters with cycle stage. These results suggest that the stages of proestrus and estrus are associated with higher seizure burden in IHKA mice. The IHKA model may thus recapitulate at least some aspects of reproductive cycle-associated seizure clustering.
Collapse
Affiliation(s)
- Jiang Li
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leanna K Leverton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Laxmi Manisha Naganatanahalli
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
18
|
Heppt J, Wittmann MT, Schäffner I, Billmann C, Zhang J, Vogt-Weisenhorn D, Prakash N, Wurst W, Taketo MM, Lie DC. β-catenin signaling modulates the tempo of dendritic growth of adult-born hippocampal neurons. EMBO J 2020; 39:e104472. [PMID: 32929771 PMCID: PMC7604596 DOI: 10.15252/embj.2020104472] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 08/06/2020] [Accepted: 08/11/2020] [Indexed: 01/07/2023] Open
Abstract
In adult hippocampal neurogenesis, stem/progenitor cells generate dentate granule neurons that contribute to hippocampal plasticity. The establishment of a morphologically defined dendritic arbor is central to the functional integration of adult‐born neurons. We investigated the role of canonical Wnt/β‐catenin signaling in dendritogenesis of adult‐born neurons. We show that canonical Wnt signaling follows a biphasic pattern, with high activity in stem/progenitor cells, attenuation in immature neurons, and reactivation during maturation, and demonstrate that this activity pattern is required for proper dendrite development. Increasing β‐catenin signaling in maturing neurons of young adult mice transiently accelerated dendritic growth, but eventually produced dendritic defects and excessive spine numbers. In middle‐aged mice, in which protracted dendrite and spine development were paralleled by lower canonical Wnt signaling activity, enhancement of β‐catenin signaling restored dendritic growth and spine formation to levels observed in young adult animals. Our data indicate that precise timing and strength of β‐catenin signaling are essential for the correct functional integration of adult‐born neurons and suggest Wnt/β‐catenin signaling as a pathway to ameliorate deficits in adult neurogenesis during aging.
Collapse
Affiliation(s)
- Jana Heppt
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marie-Theres Wittmann
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.,Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iris Schäffner
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Charlotte Billmann
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jingzhong Zhang
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Suzhou Institute of Biomedical Engineering and Technology (SIBET), Chinese Academy of Sciences, Suzhou, China
| | - Daniela Vogt-Weisenhorn
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nilima Prakash
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Dieter Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
19
|
Martín-Suárez S, Abiega O, Ricobaraza A, Hernandez-Alcoceba R, Encinas JM. Alterations of the Hippocampal Neurogenic Niche in a Mouse Model of Dravet Syndrome. Front Cell Dev Biol 2020; 8:654. [PMID: 32793597 PMCID: PMC7385077 DOI: 10.3389/fcell.2020.00654] [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: 05/04/2020] [Accepted: 07/01/2020] [Indexed: 01/24/2023] Open
Abstract
Hippocampal neurogenesis, the process by which neural stem cells (NSCs) continuously generate new neurons in the dentate gyrus (DG) of most mammals including humans, is chiefly regulated by neuronal activity. Thus, severe alterations have been found in samples from epilepsy patients and in the hippocampal neurogenic niche in mouse models of epilepsy. Reactive-like and gliogenic NSCs plus aberrant newborn neurons with altered migration, morphology, and functional properties are induced by seizures in experimental models of temporal lobe epilepsy. Hippocampal neurogenesis participates in memory and learning and in the control of anxiety and stress. It has been therefore hypothesized that part of the cognitive symptoms associated with epilepsy could be promoted by impaired hippocampal neurogenesis. We here analyze for the first time the alterations of the neurogenic niche in a novel mouse model of Dravet syndrome (DS), a genetic encephalopathy with severe epilepsy in infancy and multiple neurological comorbidities. Scn1aWT/A1783V mice, hereafter referred to as DS, carrying a heterozygous and clinically relevant SCN1A mutation (A1783V) recapitulate the disease at the genetic and phenotypic levels. We demonstrate that in the neurogenic niche of young adult DS mice there are fewer NSCs, they have impaired cell division and bear reactive-like morphology. In addition, there is significant aberrant neurogenesis. Newborn immature neurons migrate abnormally, and several morphological features are drastically changed. Thus, this study shows for the first time important modifications in hippocampal neurogenesis in DS and opens venues for further research on this topic.
Collapse
Affiliation(s)
- Soraya Martín-Suárez
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Oihane Abiega
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Ana Ricobaraza
- Gene Therapy Program CIMA, IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Rubén Hernandez-Alcoceba
- Gene Therapy Program CIMA, IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Juan Manuel Encinas
- The Neural Stem Cell and Neurogenesis Laboratory, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,IKERBASQUE, The Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
20
|
Bielefeld P, Durá I, Danielewicz J, Lucassen P, Baekelandt V, Abrous D, Encinas J, Fitzsimons C. Insult-induced aberrant hippocampal neurogenesis: Functional consequences and possible therapeutic strategies. Behav Brain Res 2019; 372:112032. [DOI: 10.1016/j.bbr.2019.112032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/29/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
|
21
|
Abstract
Compelling evidence indicates that hippocampal dentate granule cells are generated throughout human life and into old age. While animal studies demonstrate that these new neurons are important for memory function, animal research also implicates these cells in the pathogenesis of temporal lobe epilepsy. Several recent preclinical studies in rodents now suggest that targeting these new neurons can have disease-modifying effects in epilepsy.
Collapse
Affiliation(s)
- Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Anesthesia, University of Cincinnati, Cincinnati, OH, USA.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Center for Pediatric Neuroscience, Cincinnati Children's Hospital, Cincinnati, OH, USA
| |
Collapse
|
22
|
Impact of mTOR hyperactive neurons on the morphology and physiology of adjacent neurons: Do PTEN KO cells make bad neighbors? Exp Neurol 2019; 321:113029. [PMID: 31377403 DOI: 10.1016/j.expneurol.2019.113029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 01/18/2023]
Abstract
Hyperactivation of the mechanistic target of rapamycin (mTOR) pathway is associated with epilepsy, autism and brain growth abnormalities in humans. mTOR hyperactivation often results from developmental somatic mutations, producing genetic lesions and associated dysfunction in relatively restricted populations of neurons. Disrupted brain regions, such as those observed in focal cortical dysplasia, can contain a mix of normal and mutant cells. Mutant cells exhibit robust anatomical and physiological changes. Less clear, however, is whether adjacent, initially normal cells are affected by the presence of abnormal cells. To explore this question, we used a conditional, inducible mouse model approach to delete the mTOR negative regulator phosphatase and tensin homolog (PTEN) from <1% to >30% of hippocampal dentate granule cells. We then examined the morphology of PTEN-expressing granule cells located in the same dentate gyri as the knockout (KO) cells. Despite the development of spontaneous seizures in higher KO animals, and disease worsening with increasing age, the morphology and physiology of PTEN-expressing cells was only modestly affected. PTEN-expressing cells had smaller somas than cells from control animals, but other parameters were largely unchanged. These findings contrast with the behavior of PTEN KO cells, which show increasing dendritic extent with greater KO cell load. Together, the findings indicate that genetically normal neurons can exhibit relatively stable morphology and intrinsic physiology in the presence of nearby pathological neurons and systemic disease.
Collapse
|
23
|
Kalinina A, Maletta T, Carr J, Lehmann H, Fournier NM. Spatial exploration induced expression of immediate early genes Fos and Zif268 in adult-born neurons Is reduced after pentylenetetrazole kindling. Brain Res Bull 2019; 152:74-84. [PMID: 31279580 DOI: 10.1016/j.brainresbull.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 01/05/2023]
Abstract
Seizure activity stimulates adult neurogenesis, the birth of new neurons, in the hippocampus. Many new neurons that develop in the presence of repeatedly induced seizures acquire abnormal morphological and functional characteristics that can promote network hyperexcitability and hippocampal dysfunction. However, the impact of seizure induced neurogenesis on behaviour remains poorly understood. In this study, we investigated whether adult-born neurons generated immediately before and during chronic seizures were capable of integration into behaviorally relevant hippocampal networks. Adult rats underwent pentylenetetrazole (PTZ) kindling for either 1 or 2 weeks. Proliferating cells were labelled with BrdU immediately before kindling commenced. Twenty-four hours after receiving their last kindling treatment, rats were placed in a novel environment and allowed to freely explore for 30 min. The rats were euthanized 90 min later to examine for behaviourally-induced immediate early gene expression (c-fos, Zif268). Using this approach, we found that PTZ kindled rats did not differ from control rats in regards to exploratory behaviour, but there was a marked attenuation in behaviour-induced expression of Fos and Zif268 for rats that received 2 weeks of PTZ kindling. Further examination revealed that PTZ kindled rats showed reduced colocalization of Fos and Zif268 in 2.5 week old BrdU + cells. The proportion of immature granule cells (doublecortin-positive) expressing behaviorally induced Zif268 was also significantly lower for PTZ kindled rats than control rats. These results suggest that chronic seizures can potentially disrupt the ability of adult-born cells to functionally integrate into hippocampal circuits important for the processing of spatial information.
Collapse
Affiliation(s)
- Alena Kalinina
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| | - Teresa Maletta
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| | - Joshua Carr
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| | - Hugo Lehmann
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| | - Neil M Fournier
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada.
| |
Collapse
|
24
|
Ngwenya LB, Danzer SC. Impact of Traumatic Brain Injury on Neurogenesis. Front Neurosci 2019; 12:1014. [PMID: 30686980 PMCID: PMC6333744 DOI: 10.3389/fnins.2018.01014] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022] Open
Abstract
New neurons are generated in the hippocampal dentate gyrus from early development through adulthood. Progenitor cells and immature granule cells in the subgranular zone are responsive to changes in their environment; and indeed, a large body of research indicates that neuronal interactions and the dentate gyrus milieu regulates granule cell proliferation, maturation, and integration. Following traumatic brain injury (TBI), these interactions are dramatically altered. In addition to cell losses from injury and neurotransmitter dysfunction, patients often show electroencephalographic evidence of cortical spreading depolarizations and seizure activity after TBI. Furthermore, treatment for TBI often involves interventions that alter hippocampal function such as sedative medications, neuromodulating agents, and anti-epileptic drugs. Here, we review hippocampal changes after TBI and how they impact the coordinated process of granule cell adult neurogenesis. We also discuss clinical TBI treatments that have the potential to alter neurogenesis. A thorough understanding of the impact that TBI has on neurogenesis will ultimately be needed to begin to design novel therapeutics to promote recovery.
Collapse
Affiliation(s)
- Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, United States.,Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States.,Neurotrauma Center, University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, United States
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Anesthesia, University of Cincinnati, Cincinnati, OH, United States.,Center for Pediatric Neuroscience, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
25
|
Beamer EH, Jurado-Arjona J, Jimenez-Mateos EM, Morgan J, Reschke CR, Kenny A, de Leo G, Olivos-Oré LA, Arribas-Blázquez M, Madden SF, Merchán-Rubira J, Delanty N, Farrell MA, O'Brien DF, Avila J, Diaz-Hernandez M, Miras-Portugal MT, Artalejo AR, Hernandez F, Henshall DC, Engel T. MicroRNA-22 Controls Aberrant Neurogenesis and Changes in Neuronal Morphology After Status Epilepticus. Front Mol Neurosci 2018; 11:442. [PMID: 30618601 PMCID: PMC6298134 DOI: 10.3389/fnmol.2018.00442] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
Prolonged seizures (status epilepticus, SE) may drive hippocampal dysfunction and epileptogenesis, at least partly, through an elevation in neurogenesis, dysregulation of migration and aberrant dendritic arborization of newly-formed neurons. MicroRNA-22 was recently found to protect against the development of epileptic foci, but the mechanisms remain incompletely understood. Here, we investigated the contribution of microRNA-22 to SE-induced aberrant adult neurogenesis. SE was induced by intraamygdala microinjection of kainic acid (KA) to model unilateral hippocampal neuropathology in mice. MicroRNA-22 expression was suppressed using specific oligonucleotide inhibitors (antagomir-22) and newly-formed neurons were visualized using the thymidine analog iodo-deoxyuridine (IdU) and a green fluorescent protein (GFP)-expressing retrovirus to visualize the dendritic tree and synaptic spines. Using this approach, we quantified differences in the rate of neurogenesis and migration, the structure of the apical dendritic tree and density and morphology of dendritic spines in newly-formed neurons.SE resulted in an increased rate of hippocampal neurogenesis, including within the undamaged contralateral dentate gyrus (DG). Newly-formed neurons underwent aberrant migration, both within the granule cell layer and into ectopic sites. Inhibition of microRNA-22 exacerbated these changes. The dendritic diameter and the density and average volume of dendritic spines were unaffected by SE, but these parameters were all elevated in mice in which microRNA-22 was suppressed. MicroRNA-22 inhibition also reduced the length and complexity of the dendritic tree, independently of SE. These data indicate that microRNA-22 is an important regulator of morphogenesis of newly-formed neurons in adults and plays a role in supressing aberrant neurogenesis associated with SE.
Collapse
Affiliation(s)
- Edward H Beamer
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jeronimo Jurado-Arjona
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Centro Investigación Biomédica en Red Enfermedades Neurodegenerativa (CIBERNED), Madrid, Spain
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James Morgan
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Cristina R Reschke
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, RCSI, Dublin, Ireland
| | - Aidan Kenny
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gioacchino de Leo
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Luis A Olivos-Oré
- Department of Pharmacology and Toxicology, Veterinary Faculty, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Marina Arribas-Blázquez
- Department of Pharmacology and Toxicology, Veterinary Faculty, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jesús Merchán-Rubira
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Centro Investigación Biomédica en Red Enfermedades Neurodegenerativa (CIBERNED), Madrid, Spain
| | - Norman Delanty
- FutureNeuro Research Centre, RCSI, Dublin, Ireland.,Beaumont Hospital, Dublin, Ireland
| | | | | | - Jesus Avila
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Centro Investigación Biomédica en Red Enfermedades Neurodegenerativa (CIBERNED), Madrid, Spain
| | - Miguel Diaz-Hernandez
- Department of Biochemistry and Molecular Biology, Veterinary Faculty, Universidad Complutense de Madrid, Madrid, Spain
| | - M Teresa Miras-Portugal
- Department of Biochemistry and Molecular Biology, Veterinary Faculty, Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio R Artalejo
- Department of Pharmacology and Toxicology, Veterinary Faculty, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Felix Hernandez
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Centro Investigación Biomédica en Red Enfermedades Neurodegenerativa (CIBERNED), Madrid, Spain
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, RCSI, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro Research Centre, RCSI, Dublin, Ireland
| |
Collapse
|
26
|
Danzer SC. Contributions of Adult-Generated Granule Cells to Hippocampal Pathology in Temporal Lobe Epilepsy: A Neuronal Bestiary. Brain Plast 2018; 3:169-181. [PMID: 30151341 PMCID: PMC6091048 DOI: 10.3233/bpl-170056] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hippocampal neurogenesis continues throughout life in mammals – including humans. During the development of temporal lobe epilepsy, newly-generated hippocampal granule cells integrate abnormally into the brain. Abnormalities include ectopic localization of newborn cells, de novo formation of abnormal basal dendrites, and disruptions of the apical dendritic tree. Changes in granule cell position and dendritic structure fundamentally alter the types of inputs these cells are able to receive, as well as the relative proportions of remaining inputs. Dendritic abnormalities also create new pathways for recurrent excitation in the hippocampus. These abnormalities are hypothesized to contribute to the development of epilepsy, and may underlie cognitive disorders associated with the disease as well. To test this hypothesis, investigators have used pharmacological and genetic strategies in animal models to alter neurogenesis rates, or ablate the newborn cells outright. While findings are mixed and many unanswered questions remain, numerous studies now demonstrate that ablating newborn granule cells can have disease modifying effects in epilepsy. Taken together, findings provide a strong rationale for continued work to elucidate the role of newborn granule cells in epilepsy: both to understand basic mechanisms underlying the disease, and as a potential novel therapy for epilepsy.
Collapse
Affiliation(s)
- Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
27
|
Godale CM, Danzer SC. Signaling Pathways and Cellular Mechanisms Regulating Mossy Fiber Sprouting in the Development of Epilepsy. Front Neurol 2018; 9:298. [PMID: 29774009 PMCID: PMC5943493 DOI: 10.3389/fneur.2018.00298] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/17/2018] [Indexed: 02/04/2023] Open
Abstract
The sprouting of hippocampal dentate granule cell axons, termed mossy fibers, into the dentate inner molecular layer is one of the most consistent findings in tissue from patients with mesial temporal lobe epilepsy. Decades of research in animal models have revealed that mossy fiber sprouting creates de novo recurrent excitatory connections in the hippocampus, fueling speculation that the pathology may drive temporal lobe epileptogenesis. Conducting definitive experiments to test this hypothesis, however, has been challenging due to the difficulty of dissociating this sprouting from the many other changes occurring during epileptogenesis. The field has been largely driven, therefore, by correlative data. Recently, the development of powerful transgenic mouse technologies and the discovery of novel drug targets has provided new tools to assess the role of mossy fiber sprouting in epilepsy. We can now selectively manipulate hippocampal granule cells in rodent epilepsy models, providing new insights into the granule cell subpopulations that participate in mossy fiber sprouting. The cellular pathways regulating this sprouting are also coming to light, providing new targets for pharmacological intervention. Surprisingly, many investigators have found that blocking mossy fiber sprouting has no effect on seizure occurrence, while seizure frequency can be reduced by treatments that have no effect on this sprouting. These results raise new questions about the role of mossy fiber sprouting in epilepsy. Here, we will review these findings with particular regard to the contributions of new granule cells to mossy fiber sprouting and the regulation of this sprouting by the mTOR signaling pathway.
Collapse
Affiliation(s)
- Christin M Godale
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States.,Department of Anesthesia, University of Cincinnati, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
28
|
Sharma S, Puttachary S, Thippeswamy T. Glial source of nitric oxide in epileptogenesis: A target for disease modification in epilepsy. J Neurosci Res 2017; 97:1363-1377. [PMID: 29230865 DOI: 10.1002/jnr.24205] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/31/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
Epileptogenesis is the process of developing an epileptic condition and/or its progression once it is established. The molecules that initiate, promote, and propagate remarkable changes in the brain during epileptogenesis are emerging as targets for prevention/treatment of epilepsy. Epileptogenesis is a continuous process that follows immediately after status epilepticus (SE) in animal models of acquired temporal lobe epilepsy (TLE). Both SE and epileptogenesis are potential therapeutic targets for the discovery of anticonvulsants and antiepileptogenic or disease-modifying agents. For translational studies, SE targets are appropriate for screening anticonvulsive drugs prior to their advancement as therapeutic agents, while targets of epileptogenesis are relevant for identification and development of therapeutic agents that can either prevent or modify the disease or its onset. The acute seizure models do not reveal antiepileptogenic properties of anticonvulsive drugs. This review highlights the important components of epileptogenesis and the long-term impact of intervening one of these components, nitric oxide (NO), in rat and mouse kainate models of TLE. NO is a putative pleotropic gaseous neurotransmitter and an important contributor of nitro-oxidative stress that coexists with neuroinflammation and epileptogenesis. The long-term impact of inhibiting the glial source of NO during early epileptogenesis in the rat model of TLE is reviewed. The importance of sex as a biological variable in disease modification strategies in epilepsy is also briefly discussed.
Collapse
Affiliation(s)
- Shaunik Sharma
- Epilepsy Research Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| | | | - Thimmasettappa Thippeswamy
- Epilepsy Research Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| |
Collapse
|
29
|
Sherstnev VV, Kedrov AV, Solov’eva OA, Gruden’ MA, Konovalova EV, Kalinin IA, Proshin AT. The effects of α-synuclein oligomers on neurogenesis in the hippocampus and the behavior of aged mice. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417040092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
30
|
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
|
31
|
Bielefeld P, Mooney C, Henshall DC, Fitzsimons CP. miRNA-Mediated Regulation of Adult Hippocampal Neurogenesis; Implications for Epilepsy. Brain Plast 2017; 3:43-59. [PMID: 29765859 PMCID: PMC5928558 DOI: 10.3233/bpl-160036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hippocampal neural stem/progenitor cells (NSPCs) proliferate and differentiate to generate new neurons across the life span of most mammals, including humans. This process takes place within a characteristic local microenvironment where NSPCs interact with a variety of other cell types and encounter systemic regulatory factors. Within this microenvironment, cell intrinsic gene expression programs are modulated by cell extrinsic signals through complex interactions, in many cases involving short non-coding RNA molecules, such as miRNAs. Here we review the regulation of gene expression in NSPCs by miRNAs and its possible implications for epilepsy, which has been linked to alterations in adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Pascal Bielefeld
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam, The Netherlands
| | - Catherine Mooney
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David C. Henshall
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Carlos P. Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam, The Netherlands
| |
Collapse
|
32
|
Santos VR, Pun RYK, Arafa SR, LaSarge CL, Rowley S, Khademi S, Bouley T, Holland KD, Garcia-Cairasco N, Danzer SC. PTEN deletion increases hippocampal granule cell excitability in male and female mice. Neurobiol Dis 2017; 108:339-351. [PMID: 28855130 DOI: 10.1016/j.nbd.2017.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/10/2017] [Accepted: 08/26/2017] [Indexed: 02/06/2023] Open
Abstract
Deletion of the mTOR pathway inhibitor PTEN from postnatally-generated hippocampal dentate granule cells causes epilepsy. Here, we conducted field potential, whole cell recording and single cell morphology studies to begin to elucidate the mechanisms by which granule cell-specific PTEN-loss produces disease. Cells from both male and female mice were recorded to identify sex-specific effects. PTEN knockout granule cells showed altered intrinsic excitability, evident as a tendency to fire in bursts. PTEN knockout granule cells also exhibited increased frequency of spontaneous excitatory synaptic currents (sEPSCs) and decreased frequency of inhibitory currents (sIPSCs), further indicative of a shift towards hyperexcitability. Morphological studies of PTEN knockout granule cells revealed larger dendritic trees, more dendritic branches and an impairment of dendrite self-avoidance. Finally, cells from both female control and female knockout mice received more sEPSCs and more sIPSCs than corresponding male cells. Despite the difference, the net effect produced statistically equivalent EPSC/IPSC ratios. Consistent with this latter observation, extracellularly evoked responses in hippocampal slices were similar between male and female knockouts. Both groups of knockouts were abnormal relative to controls. Together, these studies reveal a host of physiological and morphological changes among PTEN knockout cells likely to underlie epileptogenic activity. SIGNIFICANCE STATEMENT Hyperactivation of the mTOR pathway is associated with numerous neurological diseases, including autism and epilepsy. Here, we demonstrate that deletion of the mTOR negative regulator, PTEN, from a subset of hippocampal dentate granule impairs dendritic patterning, increases excitatory input and decreases inhibitory input. We further demonstrate that while granule cells from female mice receive more excitatory and inhibitory input than males, PTEN deletion produces mostly similar changes in both sexes. Together, these studies provide new insights into how the relatively small number (≈200,000) of PTEN knockout granule cells instigates the development of the profound epilepsy syndrome evident in both male and female animals in this model.
Collapse
Affiliation(s)
- Victor R Santos
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Raymund Y K Pun
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Salwa R Arafa
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; University of Cincinnati, College of Pharmacy, Cincinnati, OH 45267, United States
| | - Candi L LaSarge
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Shane Rowley
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Shadi Khademi
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Tom Bouley
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Katherine D Holland
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Norberto Garcia-Cairasco
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, United States.
| |
Collapse
|
33
|
Liu TT, Feng L, Liu HF, Shu Y, Xiao B. Altered axon initial segment in hippocampal newborn neurons, associated with recurrence of temporal lobe epilepsy in rats. Mol Med Rep 2017; 16:3169-3178. [PMID: 28713955 PMCID: PMC5547972 DOI: 10.3892/mmr.2017.7017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/06/2017] [Indexed: 01/23/2023] Open
Abstract
Hippocampal neurogenesis in temporal lobe epilepsy (TLE) may result in alteration of the excitability of neurons, which contributes to spontaneous recurrent seizures. Axon initial segment (AIS) structural and functional plasticity is important in the control of neuronal excitability. It remains to be elucidated whether the plasticity of AIS occurs in hippocampal newly-generated neurons that are involved in recurrent seizures following pilocarpine-induced status epilepticus (SE). The present study first established a pilocarpine-induced TLE rat model to assess the features of newborn neurons and AIS plasticity alterations using double immunofluorescence staining of Ankyrin G and doublecortin (DCX). AIS plasticity alterations include length and distance from soma in the hippocampal newly-generated neurons post-SE. The results of the present study demonstrated that pilocarpine-induced epileptic rats exhibited aberrant hippocampal neurogenesis and longer DCX-labeled cell dendrites in the dentate gyrus. Pilocarpine-induced epileptic rats demonstrated shortened lengths of AIS and an increased distance from the soma in hippocampal newborn neurons. Mibefradil, a T/L-type calcium blocker, reversed the alterations in length and position of AIS in hippocampal newborn neurons post-SE, accompanied by decreased long-term seizure activity without increased aberrant neurogenesis. These findings indicate that the plasticity of AIS in hippocampal neurogenesis may have profound consequences in epilepsy, at least in animals.
Collapse
Affiliation(s)
- Tian-Tian Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Heng-Fang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
34
|
Ablation of Newly Generated Hippocampal Granule Cells Has Disease-Modifying Effects in Epilepsy. J Neurosci 2017; 36:11013-11023. [PMID: 27798182 DOI: 10.1523/jneurosci.1371-16.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/21/2016] [Indexed: 12/30/2022] Open
Abstract
Hippocampal granule cells generated in the weeks before and after an epileptogenic brain injury can integrate abnormally into the dentate gyrus, potentially mediating temporal lobe epileptogenesis. Previous studies have demonstrated that inhibiting granule cell production before an epileptogenic brain insult can mitigate epileptogenesis. Here, we extend upon these findings by ablating newly generated cells after the epileptogenic insult using a conditional, inducible diphtheria-toxin receptor expression strategy in mice. Diphtheria-toxin receptor expression was induced among granule cells born up to 5 weeks before pilocarpine-induced status epilepticus and these cells were then eliminated beginning 3 d after the epileptogenic injury. This treatment produced a 50% reduction in seizure frequency, but also a 20% increase in seizure duration, when the animals were examined 2 months later. These findings provide the first proof-of-concept data demonstrating that granule cell ablation therapy applied at a clinically relevant time point after injury can have disease-modifying effects in epilepsy. SIGNIFICANCE STATEMENT These findings support the long-standing hypothesis that newly generated dentate granule cells are pro-epileptogenic and contribute to the occurrence of seizures. This work also provides the first evidence that ablation of newly generated granule cells can be an effective therapy when begun at a clinically relevant time point after an epileptogenic insult. The present study also demonstrates that granule cell ablation, while reducing seizure frequency, paradoxically increases seizure duration. This paradoxical effect may reflect a disruption of homeostatic mechanisms that normally act to reduce seizure duration, but only when seizures occur frequently.
Collapse
|
35
|
Patterson KP, Barry JM, Curran MM, Singh-Taylor A, Brennan G, Rismanchi N, Page M, Noam Y, Holmes GL, Baram TZ. Enduring Memory Impairments Provoked by Developmental Febrile Seizures Are Mediated by Functional and Structural Effects of Neuronal Restrictive Silencing Factor. J Neurosci 2017; 37:3799-3812. [PMID: 28275159 PMCID: PMC5394897 DOI: 10.1523/jneurosci.3748-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022] Open
Abstract
In a subset of children experiencing prolonged febrile seizures (FSs), the most common type of childhood seizures, cognitive outcomes are compromised. However, the underlying mechanisms are unknown. Here we identified significant, enduring spatial memory problems in male rats following experimental prolonged FS (febrile status epilepticus; eFSE). Remarkably, these deficits were abolished by transient, post hoc interference with the chromatin binding of the transcriptional repressor neuron restrictive silencing factor (NRSF or REST). This transcriptional regulator is known to contribute to neuronal differentiation during development and to programmed gene expression in mature neurons. The mechanisms of the eFSE-provoked memory problems involved complex disruption of memory-related hippocampal oscillations recorded from CA1, likely resulting in part from impairments of dendritic filtering of cortical inputs as well as abnormal synaptic function. Accordingly, eFSE provoked region-specific dendritic loss in the hippocampus, and aberrant generation of excitatory synapses in dentate gyrus granule cells. Blocking NRSF transiently after eFSE prevented granule cell dysmaturation, restored a functional balance of γ-band network oscillations, and allowed treated eFSE rats to encode and retrieve spatial memories. Together, these studies provide novel insights into developing networks that underlie memory, the mechanisms by which early-life seizures influence them, and the means to abrogate the ensuing cognitive problems.SIGNIFICANCE STATEMENT Whereas seizures have been the central focus of epilepsy research, they are commonly accompanied by cognitive problems, including memory impairments that contribute to poor quality of life. These deficits often arise before the onset of spontaneous seizures, or independent from them, yet the mechanisms involved are unclear. Here, using a rodent model of common developmental seizures that provoke epilepsy in a subset of individuals, we identify serious consequent memory problems. We uncover molecular, cellular, and circuit-level mechanisms that underlie these deficits and successfully abolish them by targeted therapeutic interventions. These findings may be important for understanding and preventing cognitive problems in individuals suffering long febrile seizures.
Collapse
Affiliation(s)
| | - Jeremy M Barry
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | | | | | - Gary Brennan
- Departments of Anatomy/Neurobiology
- Pediatrics, and
| | | | - Matias Page
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | | | - Gregory L Holmes
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Tallie Z Baram
- Departments of Anatomy/Neurobiology,
- Pediatrics, and
- Neurology, University of California-Irvine, Irvine, California 92697-4475, and
| |
Collapse
|
36
|
Kim SR. Control of Granule Cell Dispersion by Natural Materials Such as Eugenol and Naringin: A Potential Therapeutic Strategy Against Temporal Lobe Epilepsy. J Med Food 2016; 19:730-6. [PMID: 27404051 DOI: 10.1089/jmf.2016.3712] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The hippocampus is an important brain area where abnormal morphological characteristics are often observed in patients with temporal lobe epilepsy (TLE), typically showing the loss of the principal neurons in the CA1 and CA3 areas of the hippocampus. TLE is frequently associated with widening of the granule cell layer of the dentate gyrus (DG), termed granule cell dispersion (GCD), in the hippocampus, suggesting that the control of GCD with protection of hippocampal neurons may be useful for preventing and inhibiting epileptic seizures. We previously reported that eugenol (EUG), which is an essential component of medicinal herbs and has anticonvulsant activity, is beneficial for treating epilepsy through its ability to inhibit GCD via suppression of mammalian target of rapamycin complex 1 (mTORC1) activation in the hippocampal DG in a kainic acid (KA)-treated mouse model of epilepsy in vivo. In addition, we reported that naringin, a bioflavonoid in citrus fruits, could exert beneficial effects, such as antiautophagic stress and antineuroinflammation, in the KA mouse model of epilepsy, even though it was unclear whether naringin might also attenuate the seizure-induced morphological changes of GCD in the DG. Similar to the effects of EUG, we recently observed that naringin treatment significantly reduced KA-induced GCD and mTORC1 activation, which are both involved in epileptic seizures, in the hippocampus of mouse brain. Therefore, these observations suggest that the utilization of natural materials, which have beneficial properties such as inhibition of GCD formation and protection of hippocampal neurons, may be useful in developing a novel therapeutic agent against TLE.
Collapse
Affiliation(s)
- Sang Ryong Kim
- 1 School of Life Sciences, Kyungpook National University , Daegu, Korea.,2 BK21 plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Korea.,3 Brain Science and Engineering Institute, Kyungpook National University , Daegu, Korea
| |
Collapse
|
37
|
Naringin attenuates granule cell dispersion in the dentate gyrus in a mouse model of temporal lobe epilepsy. Epilepsy Res 2016; 123:6-10. [DOI: 10.1016/j.eplepsyres.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/18/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023]
|
38
|
Wulsin AC, Solomon MB, Privitera MD, Danzer SC, Herman JP. Hypothalamic-pituitary-adrenocortical axis dysfunction in epilepsy. Physiol Behav 2016; 166:22-31. [PMID: 27195458 DOI: 10.1016/j.physbeh.2016.05.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/04/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
Abstract
Epilepsy is a common neurological disease, affecting 2.4million people in the US. Among the many different forms of the disease, temporal lobe epilepsy (TLE) is one of the most frequent in adults. Recent studies indicate the presence of a hyperactive hypothalamopituitary- adrenocortical (HPA) axis and elevated levels of glucocorticoids in TLE patients. Moreover, in these patients, stress is a commonly reported trigger of seizures, and stress-related psychopathologies, including depression and anxiety, are highly prevalent. Elevated glucocorticoids have been implicated in the development of stress-related psychopathologies. Similarly, excess glucocorticoids have been found to increase neuronal excitability, epileptiform activity and seizure susceptibility. Thus, patients with TLE may generate abnormal stress responses that both facilitate ictal discharges and increase vulnerability for the development of comorbid psychopathologies. Here, we will examine the evidence that the HPA axis is disrupted in TLE, consider potential mechanisms by which this might occur, and discuss the implications of HPA dysfunction for seizuretriggering and psychiatric comorbidities.
Collapse
Affiliation(s)
- Aynara C Wulsin
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Department of Anesthesia, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States.
| | - Matia B Solomon
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Michael D Privitera
- Department of Neurology, Neuroscience Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Steve C Danzer
- Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Department of Anesthesia, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
39
|
Impact of rapamycin on status epilepticus induced hippocampal pathology and weight gain. Exp Neurol 2016; 280:1-12. [PMID: 26995324 DOI: 10.1016/j.expneurol.2016.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Growing evidence implicates the dentate gyrus in temporal lobe epilepsy (TLE). Dentate granule cells limit the amount of excitatory signaling through the hippocampus and exhibit striking neuroplastic changes that may impair this function during epileptogenesis. Furthermore, aberrant integration of newly-generated granule cells underlies the majority of dentate restructuring. Recently, attention has focused on the mammalian target of rapamycin (mTOR) signaling pathway as a potential mediator of epileptogenic change. Systemic administration of the mTOR inhibitor rapamycin has promising therapeutic potential, as it has been shown to reduce seizure frequency and seizure severity in rodent models. Here, we tested whether mTOR signaling facilitates abnormal development of granule cells during epileptogenesis. We also examined dentate inflammation and mossy cell death in the dentate hilus. To determine if mTOR activation is necessary for abnormal granule cell development, transgenic mice that harbored fluorescently-labeled adult-born granule cells were treated with rapamycin following pilocarpine-induced status epilepticus. Systemic rapamycin effectively blocked phosphorylation of S6 protein (a readout of mTOR activity) and reduced granule cell mossy fiber axon sprouting. However, the accumulation of ectopic granule cells and granule cells with aberrant basal dendrites was not significantly reduced. Mossy cell death and reactive astrocytosis were also unaffected. These data suggest that anti-epileptogenic effects of mTOR inhibition may be mediated by mechanisms other than inhibition of these common dentate pathologies. Consistent with this conclusion, rapamycin prevented pathological weight gain in epileptic mice, suggesting that rapamycin might act on central circuits or even peripheral tissues controlling weight gain in epilepsy.
Collapse
|
40
|
Pineda JR, Encinas JM. The Contradictory Effects of Neuronal Hyperexcitation on Adult Hippocampal Neurogenesis. Front Neurosci 2016; 10:74. [PMID: 26973452 PMCID: PMC4776215 DOI: 10.3389/fnins.2016.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/17/2016] [Indexed: 01/19/2023] Open
Abstract
Adult hippocampal neurogenesis is a highly plastic process that responds swiftly to neuronal activity. Adult hippocampal neurogenesis can be regulated at the level of neural stem cell recruitment and activation, progenitor proliferation, as well as newborn cell survival and differentiation. An "excitation-neurogenesis" rule was proposed after the demonstration of the capability of cultured neural stem and progenitor cells to intrinsically sense neuronal excitatory activity. In vivo, this property has remained elusive although recently the direct response of neural stem cells to GABA in the hippocampus via GABAA receptors has evidenced a mechanism for a direct talk between neurons and neural stem cells. As it is pro-neurogenic, the effect of excitatory neuronal activity has been generally considered beneficial. But what happens in situations of neuronal hyperactivity in which neurogenesis can be dramatically boosted? In animal models, electroconvulsive shock markedly increases neurogenesis. On the contrary, in epilepsy rodent models, seizures induce the generation of misplaced neurons with abnormal morphological and electrophysiological properties, namely aberrant neurogenesis. We will herein discuss what is known about the mechanisms of influence of neurons on neural stem cells, as well as the severe effects of neuronal hyperexcitation on hippocampal neurogenesis.
Collapse
Affiliation(s)
- José R Pineda
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience Zamudio, Spain
| | - Juan M Encinas
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for NeuroscienceZamudio, Spain; IKERBASQUE, The Basque Foundation for ScienceBilbao, Spain
| |
Collapse
|
41
|
Scharfman HE, Myers CE. Corruption of the dentate gyrus by "dominant" granule cells: Implications for dentate gyrus function in health and disease. Neurobiol Learn Mem 2016; 129:69-82. [PMID: 26391451 PMCID: PMC4792754 DOI: 10.1016/j.nlm.2015.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/31/2022]
Abstract
The dentate gyrus (DG) and area CA3 of the hippocampus are highly organized lamellar structures which have been implicated in specific cognitive functions such as pattern separation and pattern completion. Here we describe how the anatomical organization and physiology of the DG and CA3 are consistent with structures that perform pattern separation and completion. We then raise a new idea related to the complex circuitry of the DG and CA3 where CA3 pyramidal cell 'backprojections' play a potentially important role in the sparse firing of granule cells (GCs), considered important in pattern separation. We also propose that GC axons, the mossy fibers, already known for their highly specialized structure, have a dynamic function that imparts variance--'mossy fiber variance'--which is important to pattern separation and completion. Computational modeling is used to show that when a subset of GCs become 'dominant,' one consequence is loss of variance in the activity of mossy fiber axons and a reduction in pattern separation and completion in the model. Empirical data are then provided using an example of 'dominant' GCs--subsets of GCs that develop abnormally and have increased excitability. Notably, these abnormal GCs have been identified in animal models of disease where DG-dependent behaviors are impaired. Together these data provide insight into pattern separation and completion, and suggest that behavioral impairment could arise from dominance of a subset of GCs in the DG-CA3 network.
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, United States; Departments of Child & Adolescent Psychiatry, Physiology & Neuroscience, and Psychiatry, New York University Langone Medical Center, United States.
| | - Catherine E Myers
- VA New Jersey Health Care System, VA Medical Center, NeuroBehavioral Research Lab (Mail Stop 15a), 385 Tremont Avenue, East Orange, NJ 07018, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, United States
| |
Collapse
|
42
|
Korn MJ, Mandle QJ, Parent JM. Conditional Disabled-1 Deletion in Mice Alters Hippocampal Neurogenesis and Reduces Seizure Threshold. Front Neurosci 2016; 10:63. [PMID: 26941603 PMCID: PMC4766299 DOI: 10.3389/fnins.2016.00063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/10/2016] [Indexed: 11/13/2022] Open
Abstract
Many animal models of temporal lobe epilepsy (TLE) exhibit altered neurogenesis arising from progenitors within the dentate gyrus subgranular zone (SGZ). Aberrant integration of new neurons into the existing circuit is thought to contribute to epileptogenesis. In particular, adult-born neurons that exhibit ectopic migration and hilar basal dendrites (HBDs) are suggested to be pro-epileptogenic. Loss of reelin signaling may contribute to these morphological changes in patients with epilepsy. We previously demonstrated that conditional deletion of the reelin adaptor protein, disabled-1 (Dab1), from postnatal mouse SGZ progenitors generated dentate granule cells (DGCs) with abnormal dendritic development and ectopic placement. To determine whether the early postnatal loss of reelin signaling is epileptogenic, we conditionally deleted Dab1 in neural progenitors and their progeny on postnatal days 7–8 and performed chronic video-EEG recordings 8–10 weeks later. Dab1-deficient mice did not have spontaneous seizures but exhibited interictal epileptiform abnormalities and a significantly reduced latency to pilocarpine-induced status epilepticus. After chemoconvulsant treatment, over 90% of mice deficient for Dab1 developed generalized motor convulsions with tonic-clonic movements, rearing, and falling compared to <20% of wild-type mice. Recombination efficiency, measured by Cre reporter expression, inversely correlated with time to the first sustained seizure. These pro-epileptogenic changes were associated with decreased neurogenesis and increased numbers of hilar ectopic DGCs. Interestingly, neurons co-expressing the Cre reporter comprised a fraction of these hilar ectopic DGCs cells, suggesting a non-cell autonomous effect for the loss of reelin signaling. We also noted a dispersion of the CA1 pyramidal layer, likely due to hypomorphic effects of the conditional Dab1 allele, but this abnormality did not correlate with seizure susceptibility. These findings suggest that the misplacement or reduction of postnatally-generated DGCs contributes to aberrant circuit development and hyperexcitability, but aberrant neurogenesis after conditional Dab1 deletion alone is not sufficient to produce spontaneous seizures.
Collapse
Affiliation(s)
- Matthew J Korn
- Department of Neurology, University of Michigan Medical Center Ann Arbor, MI, USA
| | - Quinton J Mandle
- Department of Neurology, University of Michigan Medical Center Ann Arbor, MI, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan Medical CenterAnn Arbor, MI, USA; VA Ann Arbor Healthcare SystemAnn Arbor, MI, USA
| |
Collapse
|
43
|
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
|
44
|
Gao F, Song X, Zhu D, Wang X, Hao A, Nadler JV, Zhan RZ. Dendritic morphology, synaptic transmission, and activity of mature granule cells born following pilocarpine-induced status epilepticus in the rat. Front Cell Neurosci 2015; 9:384. [PMID: 26500490 PMCID: PMC4596052 DOI: 10.3389/fncel.2015.00384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 09/14/2015] [Indexed: 01/23/2023] Open
Abstract
To understand the potential role of enhanced hippocampal neurogenesis after pilocarpine-induced status epilepticus (SE) in the development of epilepsy, we quantitatively analyzed the geometry of apical dendrites, synaptic transmission, and activation levels of normotopically distributed mature newborn granule cells in the rat. SE in male Sprague-Dawley rats (between 6 and 7 weeks old) lasting for more than 2 h was induced by an intraperitoneal injection of pilocarpine. The complexity, spine density, miniature post-synaptic currents, and activity-regulated cytoskeleton-associated protein (Arc) expression of granule cells born 5 days after SE were studied between 10 and 17 weeks after CAG-GFP retroviral vector-mediated labeling. Mature granule cells born after SE had dendritic complexity similar to that of granule cells born naturally, but with denser mushroom-like spines in dendritic segments located in the outer molecular layer. Miniature inhibitory post-synaptic currents (mIPSCs) were similar between the controls and rats subjected to SE; however, smaller miniature excitatory post-synaptic current (mEPSC) amplitude with a trend toward less frequent was found in mature granule cells born after SE. After maturation, granule cells born after SE did not show denser Arc expression in the resting condition or 2 h after being activated by pentylenetetrazol-induced transient seizure activity than vicinal GFP-unlabeled granule cells. Thus our results suggest that normotopic granule cells born after pilocarpine-induced SE are no more active when mature than age-matched, naturally born granule cells.
Collapse
Affiliation(s)
- Fei Gao
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Xueying Song
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Dexiao Zhu
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Xiaochen Wang
- Department of Physiology, Shandong University School of Medicine Jinan, China
| | - Aijun Hao
- Department of Histology and Embryology, Shandong University School of Medicine Jinan, China
| | - J Victor Nadler
- Departments of Pharmacology and Neurobiology, Duke University Medical Center Durham, NC, USA
| | - Ren-Zhi Zhan
- Department of Physiology, Shandong University School of Medicine Jinan, China
| |
Collapse
|
45
|
Jeong KH, Lee DS, Kim SR. Effects of eugenol on granule cell dispersion in a mouse model of temporal lobe epilepsy. Epilepsy Res 2015. [DOI: 10.1016/j.eplepsyres.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
46
|
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
|
47
|
LaSarge CL, Santos VR, Danzer SC. PTEN deletion from adult-generated dentate granule cells disrupts granule cell mossy fiber axon structure. Neurobiol Dis 2015; 75:142-50. [PMID: 25600212 PMCID: PMC4351143 DOI: 10.1016/j.nbd.2014.12.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/17/2014] [Accepted: 12/26/2014] [Indexed: 01/30/2023] Open
Abstract
Dysregulation of the mTOR-signaling pathway is implicated in the development of temporal lobe epilepsy. In mice, deletion of PTEN from hippocampal dentate granule cells leads to mTOR hyperactivation and promotes the rapid onset of spontaneous seizures. The mechanism by which these abnormal cells initiate epileptogenesis, however, is unclear. PTEN-knockout granule cells develop abnormally, exhibiting morphological features indicative of increased excitatory input. If these cells are directly responsible for seizure genesis, it follows that they should also possess increased output. To test this prediction, dentate granule cell axon morphology was quantified in control and PTEN-knockout mice. Unexpectedly, PTEN deletion increased giant mossy fiber bouton spacing along the axon length, suggesting reduced innervation of CA3. Increased width of the mossy fiber axon pathway in stratum lucidum, however, which likely reflects an unusual increase in mossy fiber axon collateralization in this region, offsets the reduction in boutons per axon length. These morphological changes predict a net increase in granule cell innervation of CA3. Increased diameter of axons from PTEN-knockout cells would further enhance granule cell communication with CA3. Altogether, these findings suggest that amplified information flow through the hippocampal circuit contributes to seizure occurrence in the PTEN-knockout mouse model of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Candi L LaSarge
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Victor R Santos
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Physiology, Ribeirão Preto Medical School, Ribeirão Preto, SP, Brazil
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesia, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
48
|
Song C, Xu W, Zhang X, Wang S, Zhu G, Xiao T, Zhao M, Zhao C. CXCR4 Antagonist AMD3100 Suppresses the Long-Term Abnormal Structural Changes of Newborn Neurons in the Intraventricular Kainic Acid Model of Epilepsy. Mol Neurobiol 2015; 53:1518-1532. [PMID: 25650120 DOI: 10.1007/s12035-015-9102-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/15/2015] [Indexed: 12/19/2022]
Abstract
Abnormal hippocampal neurogenesis is a prominent feature of temporal lobe epilepsy (TLE) models, which is thought to contribute to abnormal brain activity. Stromal cell-derived factor-1 (SDF-1) and its specific receptor CXCR4 play important roles in adult neurogenesis. We investigated whether treatment with the CXCR4 antagonist AMD3100 suppressed aberrant hippocampal neurogenesis, as well as the long-term consequences in the intracerebroventricular kainic acid (ICVKA) model of epilepsy. Adult male rats were randomly assigned as control rats, rats subjected to status epilepticus (SE), and post-SE rats treated with AMD3100. Animals in each group were divided into two subgroups (acute stage and chronic stage). We used immunofluorescence staining of BrdU and DCX to analyze the hippocampal neurogenesis on post-SE days 10 or 74. Nissl staining and Timm staining were used to evaluate hippocampal damage and mossy fiber sprouting, respectively. On post-SE day 72, the frequency and mean duration of spontaneous seizures were measured by electroencephalography (EEG). Cognitive function was evaluated by Morris water maze testing on post-SE day 68. The ICVKA model of TLE resulted in aberrant neurogenesis such as altered proliferation, abnormal dendrite development of newborn neurons, as well as spontaneous seizures and spatial learning impairments. More importantly, AMD3100 treatment reversed the aberrant neurogenesis seen after TLE, which was accompanied by decreased long-term seizure activity, though improvement in spatial learning was not seen. AMD3100 could suppress long-term seizure activity and alter adult neurogenesis in the ICVKA model of TLE, which provided morphological evidences that AMD3100 might be beneficial for treating chronic epilepsy.
Collapse
Affiliation(s)
- Chengguang Song
- Department of Neurology, The First Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China.,Department of Neurology, Benxi Central Hospital of China Medical University, Benxi, Liaoning, People's Republic of China
| | - Wangshu Xu
- Department of Neurology, The First Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaoqian Zhang
- Department of Neurology, The First Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Shang Wang
- Department of Neurology, The First Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Ting Xiao
- Department of Dermatology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China.,Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, Liaoning, People's Republic of China
| | - Mei Zhao
- Department of Cardiology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chuansheng Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, No. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
49
|
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
|
50
|
Hester MS, Danzer SC. Hippocampal granule cell pathology in epilepsy - a possible structural basis for comorbidities of epilepsy? Epilepsy Behav 2014; 38:105-16. [PMID: 24468242 PMCID: PMC4110172 DOI: 10.1016/j.yebeh.2013.12.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/17/2013] [Accepted: 12/21/2013] [Indexed: 01/31/2023]
Abstract
Temporal lobe epilepsy in both animals and humans is characterized by abnormally integrated hippocampal dentate granule cells. Among other abnormalities, these cells make axonal connections with inappropriate targets, grow dendrites in the wrong direction, and migrate to ectopic locations. These changes promote the formation of recurrent excitatory circuits, leading to the appealing hypothesis that these abnormal cells may by epileptogenic. While this hypothesis has been the subject of intense study, less attention has been paid to the possibility that abnormal granule cells in the epileptic brain may also contribute to comorbidities associated with the disease. Epilepsy is associated with a variety of general findings, such as memory disturbances and cognitive dysfunction, and is often comorbid with a number of other conditions, including schizophrenia and autism. Interestingly, recent studies implicate disruption of common genes and gene pathways in all three diseases. Moreover, while neuropsychiatric conditions are associated with changes in a variety of brain regions, granule cell abnormalities in temporal lobe epilepsy appear to be phenocopies of granule cell deficits produced by genetic mouse models of autism and schizophrenia, suggesting that granule cell dysmorphogenesis may be a common factor uniting these seemingly diverse diseases. Disruption of common signaling pathways regulating granule cell neurogenesis may begin to provide mechanistic insight into the cooccurrence of temporal lobe epilepsy and cognitive and behavioral disorders.
Collapse
Affiliation(s)
- Michael S Hester
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesia, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45267, USA; Molecular and Developmental Biology Graduate Program, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|