1
|
Scharfman HE. Adenosine Makes a Scene. Epilepsy Curr 2025:15357597251323126. [PMID: 40256115 PMCID: PMC12003313 DOI: 10.1177/15357597251323126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025] Open
Abstract
Retrograde Adenosine/A2A Receptor Signaling Facilitates Excitatory Synaptic Transmission and Seizures Nasrallah K, Berthoux C, Hashimotodani Y, Chávez AE, Gulfo MC, Luján R, Castillo PE. Cell Rep . 2024; 43(7):114382. doi: 10.1016/j.celrep.2024.114382. Retrograde signaling at the synapse is a fundamental way by which neurons communicate and neuronal circuit function is fine-tuned upon activity. While long-term changes in neurotransmitter release commonly rely on retrograde signaling, the mechanisms remain poorly understood. Here, we identified adenosine/A2A receptor (A2AR) as a retrograde signaling pathway underlying presynaptic long-term potentiation (LTP) at a hippocampal excitatory circuit critically involved in memory and epilepsy. Transient burst activity of a single dentate granule cell induced LTP of mossy cell synaptic inputs, a BDNF/TrkB-dependent form of plasticity that facilitates seizures. Postsynaptic TrkB activation released adenosine from granule cells, uncovering a non-conventional BDNF/TrkB signaling mechanism. Moreover, presynaptic A2ARs were necessary and sufficient for LTP. Lastly, seizure induction released adenosine in a TrkB-dependent manner, while removing A2ARs or TrkB from the dentate gyrus had anti-convulsant effects. By mediating presynaptic LTP, adenosine/A2AR retrograde signaling may modulate dentate gyrus-dependent learning and promote epileptic activity.
Collapse
Affiliation(s)
- Helen E Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Grossman School of Medicine and the Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research
| |
Collapse
|
2
|
Kebede V, Di Sapia R, Tonesi N, Rizzi M, Balosso S, Spallaci D, Craparotta I, Pasetto L, Bonetto V, Marsella G, Porcu L, Rosati G, Ieraci A, Vezzani A. Voluntary running wheel activity reduces seizure burden and affords neuroprotection in a mouse model of acquired epilepsy. Epilepsia 2025. [PMID: 39982429 DOI: 10.1111/epi.18313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE Physical exercise may improve neurological deficits and neuronal damage after acute brain injuries and decrease established seizures. We investigated whether voluntary running wheel (RW) activity affects epileptogenesis in a mouse model of acquired epilepsy compared to sedentary mice. METHODS Epilepsy was induced by intra-amygdala kainate causing status epilepticus (SE) in adult male mice. Sham mice were implanted with electrodes and injected with saline, and matched to experimental mice. In the RW-1 protocol, SE mice were trained to run for 5 weeks before SE induction and for 6 weeks thereafter. In the RW-2 protocol, mice began using RWs 24 h post-SE for 10 weeks. At the end of each protocol, electrocorticography (ECoG) was recorded for 2 weeks (24/7) in the absence of RWs. Matched sedentary mice were kept in home cages without RWs, subjected to SE, and had ECoG monitored. At the end of experiment, all mice were processed for assessing hippocampal neuronal cell loss (Nissl staining), hilar mossy cells (GLUR2/3 staining), and blood-brain barrier (BBB) damage (serum matrix metalloproteinase-9 [MMP-9] by enzyme-linked immunosorbent assay). Neuroinflammation (reverse-transcriptase quantitative polymerase chain reaction) and albumin level (western blot) were also measured in the hippocampus of RW1 mice 72 h post-SE, together with serum MMP-9. RESULTS RW activity in SE mice reduced the incidence of epilepsy (RW-1 by 38%; RW-2 by 54%, p < .05) and the total time spent in seizures (RW-1, p < .05; RW-2, p < .01) compared to sedentary mice. RW-1 SE mice showed reduced average seizure duration (p < .01), whereas RW-2 SE mice showed reduced number of seizures (p < .01). Reduction in seizure duration was associated with prevention of GluR2/3-positive mossy cell loss, which occurs in sedentary SE mice (p < .01 vs sham mice). Seizure duration in epileptic mice was negatively correlated with the number of hilar mossy cells (p < .01). Preventive RW-1 activity reduced SE duration and severity (p < .05) vs sedentary mice. Aberrant neurogenesis was reduced in the dentate gyrus of SE mice subjected to RWs (p < .01) vs sedentary mice. Serum MMP-9 and brain albumin levels were reduced in SE mice exposed to running activity (p < .05) compared to sedentary mice. SIGNIFICANCE Physical exercise reduced seizure burden and neuropathology in mice, offering a strategy to improve disease outcomes after acute brain injury.
Collapse
Affiliation(s)
- Valentina Kebede
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Nicole Tonesi
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Massimo Rizzi
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Silvia Balosso
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daniele Spallaci
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Craparotta
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Pasetto
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Valentina Bonetto
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gerardo Marsella
- Animal Welfare, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Porcu
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
3
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. eLife 2024; 12:RP90893. [PMID: 39446467 PMCID: PMC11501206 DOI: 10.7554/elife.90893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2Baxfl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild-type females or males, which is notable because loss of these two hilar cell types is implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, University of SaskatchewanSaskatoonCanada
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Neuroscience & Physiology, Psychiatry, and the Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
4
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.08.548217. [PMID: 37502909 PMCID: PMC10369878 DOI: 10.1101/2023.07.08.548217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2 Bax fl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were the more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild type females or males, which is notable because these two hilar cell types are implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, The University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
- Departments of Neuroscience & Physiology, Psychiatry, and the New York University, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
5
|
Ravizza T, Scheper M, Di Sapia R, Gorter J, Aronica E, Vezzani A. mTOR and neuroinflammation in epilepsy: implications for disease progression and treatment. Nat Rev Neurosci 2024; 25:334-350. [PMID: 38531962 DOI: 10.1038/s41583-024-00805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Epilepsy remains a major health concern as anti-seizure medications frequently fail, and there is currently no treatment to stop or prevent epileptogenesis, the process underlying the onset and progression of epilepsy. The identification of the pathological processes underlying epileptogenesis is instrumental to the development of drugs that may prevent the generation of seizures or control pharmaco-resistant seizures, which affect about 30% of patients. mTOR signalling and neuroinflammation have been recognized as critical pathways that are activated in brain cells in epilepsy. They represent a potential node of biological convergence in structural epilepsies with either a genetic or an acquired aetiology. Interventional studies in animal models and clinical studies give strong support to the involvement of each pathway in epilepsy. In this Review, we focus on available knowledge about the pathophysiological features of mTOR signalling and the neuroinflammatory brain response, and their interactions, in epilepsy. We discuss mitigation strategies for each pathway that display therapeutic effects in experimental and clinical epilepsy. A deeper understanding of these interconnected molecular cascades could enhance our strategies for managing epilepsy. This could pave the way for new treatments to fill the gaps in the development of preventative or disease-modifying drugs, thus overcoming the limitations of current symptomatic medications.
Collapse
Affiliation(s)
- Teresa Ravizza
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Mirte Scheper
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Jan Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands.
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy.
| |
Collapse
|
6
|
Alavi MS, Al-Asady AM, Fanoudi S, Sadeghnia HR. Differential effects of antiseizure medications on neurogenesis: Evidence from cells to animals. Heliyon 2024; 10:e26650. [PMID: 38420427 PMCID: PMC10901100 DOI: 10.1016/j.heliyon.2024.e26650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Neurogenesis, the process of generating functionally integrated neurons from neural stem and progenitor cells, is involved in brain development during embryonic stages but continues throughout life. Adult neurogenesis plays essential roles in many brain functions such as cognition, brain plasticity, and repair. Abnormalities in neurogenesis have been described in many neuropsychiatric and neurological disorders, including epilepsy. While sharing a common property of suppressing seizures, accumulating evidence has shown that some antiseizure medications (ASM) exhibit neuroprotective potential in the non-epileptic models including Parkinson's disease, Alzheimer's disease, cerebral ischemia, or traumatic brain injury. ASM are a heterogeneous group of medications with different mechanisms of actions. Therefore, it remains to be revealed whether neurogenesis is a class effect or related to them all. In this comprehensive literature study, we reviewed the literature data on the influence of ASM on the neurogenesis process during brain development and also in the adult brain under physiological or pathological conditions. Meanwhile, we discussed the underlying mechanisms associated with the neurogenic effects of ASM by linking the reported in vivo and in vitro studies. PubMed, Web of Science, and Google Scholar databases were searched until the end of February 2023. A total of 83 studies were used finally. ASM can modulate neurogenesis through the increase or decrease of proliferation, survival, and differentiation of the quiescent NSC pool. The present article indicated that the neurogenic potential of ASM depends on the administered dose, treatment period, temporal administration of the drug, and normal or disease context.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Sciences, Faculty of Nursing, University of Warith Al-Anbiyaa, Karbala, Iraq
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Karbala, Iraq
| | - Sahar Fanoudi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Hamid R Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Musaelyan K, Horowitz MA, McHugh S, Szele FG. Fluoxetine Can Cause Epileptogenesis and Aberrant Neurogenesis in Male Wild-Type Mice. Dev Neurosci 2023; 46:158-166. [PMID: 37302394 DOI: 10.1159/000531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Antidepressants in general, and fluoxetine in particular, increase adult hippocampal neurogenesis (AHN) in mice. Here we asked how the antidepressant fluoxetine affects behavior and AHN in a corticosterone model of depression. In three groups of adult male C57BL/6j mice, we administered either vehicle (VEH), corticosterone (CORT) treatment to induce a depression-like state, or corticosterone plus a standard dose of fluoxetine (CORT+FLX). Following treatment, mice performed the open field test, the novelty suppressed feeding (NSF) test, and the splash test. Neurogenesis was assessed by means of immunohistochemistry using BrdU and neuronal maturation markers. Unexpectedly, 42% of the CORT+FLX-treated mice exhibited severe weight loss, seizures, and sudden death. As expected, the CORT-treated group had altered behaviors compared to the VEH group, but the CORT+FLX mice that survived did not show any behavioral improvement compared to the CORT group. Antidepressants generally increase neurogenesis and here we also found that compared to CORT mice, CORT+FLX mice that survived had a significantly greater density of BrdU+, BrdU+DCX+, and BrdU+NeuN+ cells, suggesting increased neurogenesis. Moreover, the density of BrdU+NeuN+ cells was increased in an aberrant location, the hilus, of CORT+FLX mice, similar to previous studies describing aberrant neurogenesis following seizures. In conclusion, fluoxetine could induce considerable adverse effects in wild-type mice, including seizure-like activity. Fluoxetine-induced neurogenesis increases could be related to this activity; therefore, proneurogenic effects of fluoxetine and other antidepressants, especially in the absence of any behavioral therapeutic effects, should be interpreted with caution.
Collapse
Affiliation(s)
- Ksenia Musaelyan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Mark A Horowitz
- Research and Development Department, North East London NHS Foundation Trust, Ilford, UK
- Department of Psychiatry, University College London, London, UK
| | - Stephen McHugh
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Song B, Kim CH. Cell-autonomous PLCβ1 modulation of neural stem/progenitor cell proliferation during adult hippocampal neurogenesis. Neurosci Lett 2022; 791:136899. [DOI: 10.1016/j.neulet.2022.136899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/14/2022] [Accepted: 09/30/2022] [Indexed: 10/31/2022]
|
9
|
Beesley S, Sullenberger T, Ailani R, D'Orio C, Crockett MS, Kumar SS. d-Serine Intervention In The Medial Entorhinal Area Alters TLE-Related Pathology In CA1 Hippocampus Via The Temporoammonic Pathway. Neuroscience 2021; 453:168-186. [PMID: 33197499 PMCID: PMC7796904 DOI: 10.1016/j.neuroscience.2020.10.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 01/15/2023]
Abstract
Entrainment of the hippocampus by the medial entorhinal area (MEA) in Temporal Lobe Epilepsy (TLE), the most common type of drug-resistant epilepsy in adults, is believed to be mediated primarily through the perforant pathway (PP), which connects stellate cells in layer (L) II of the MEA with granule cells of the dentate gyrus (DG) to drive the hippocampal tri-synaptic circuit. Using immunohistochemistry, high-resolution confocal microscopy and the rat pilocarpine model of TLE, we show here that the lesser known temporoammonic pathway (TAP) plays a significant role in transferring MEA pathology to the CA1 region of the hippocampus independently of the PP. The pathology observed was region-specific and restricted primarily to the CA1c subfield of the hippocampus. As shown previously, daily intracranial infusion of d-serine (100 μm), an antagonist of GluN3-containing triheteromeric N-Methyl d-aspartate receptors (t-NMDARs), into the MEA prevented loss of LIII neurons and epileptogenesis. This intervention in the MEA led to the rescue of hippocampal CA1 neurons that would have otherwise perished in the epileptic animals, and down regulation of the expression of astrocytes and microglia thereby mitigating the effects of neuroinflammation. Interestingly, these changes were not observed to a similar extent in other regions of vulnerability like the hilus, DG or CA3, suggesting that the pathology manifest in CA1 is driven predominantly through the TAP. This work highlights TAP's role in the entrainment of the hippocampus and identifies specific areas for therapeutic intervention in dealing with TLE.
Collapse
Affiliation(s)
- Stephen Beesley
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States
| | - Thomas Sullenberger
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States
| | - Roshan Ailani
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States
| | - Cameron D'Orio
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States
| | - Mathew S Crockett
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States
| | - Sanjay S Kumar
- Department of Biomedical Sciences, College of Medicine & Program in Neuroscience, Florida State University, 1115 W. Call Street, Tallahassee, FL 32306-4300, United States.
| |
Collapse
|
10
|
Jain S, LaFrancois JJ, Botterill JJ, Alcantara-Gonzalez D, Scharfman HE. Adult neurogenesis in the mouse dentate gyrus protects the hippocampus from neuronal injury following severe seizures. Hippocampus 2019; 29:683-709. [PMID: 30672046 PMCID: PMC6640126 DOI: 10.1002/hipo.23062] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/29/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023]
Abstract
Previous studies suggest that reducing the numbers of adult-born neurons in the dentate gyrus (DG) of the mouse increases susceptibility to severe continuous seizures (status epilepticus; SE) evoked by systemic injection of the convulsant kainic acid (KA). However, it was not clear if the results would be the same for other ways to induce seizures, or if SE-induced damage would be affected. Therefore, we used pilocarpine, which induces seizures by a different mechanism than KA. Also, we quantified hippocampal damage after SE. In addition, we used both loss-of-function and gain-of-function methods in adult mice. We hypothesized that after loss-of-function, mice would be more susceptible to pilocarpine-induced SE and SE-associated hippocampal damage, and after gain-of-function, mice would be more protected from SE and hippocampal damage after SE. For loss-of-function, adult neurogenesis was suppressed by pharmacogenetic deletion of dividing radial glial precursors. For gain-of-function, adult neurogenesis was increased by conditional deletion of pro-apoptotic gene Bax in Nestin-expressing progenitors. Fluoro-Jade C (FJ-C) was used to quantify neuronal injury and video-electroencephalography (video-EEG) was used to quantify SE. Pilocarpine-induced SE was longer in mice with reduced adult neurogenesis, SE had more power and neuronal damage was greater. Conversely, mice with increased adult-born neurons had shorter SE, SE had less power, and there was less neuronal damage. The results suggest that adult-born neurons exert protective effects against SE and SE-induced neuronal injury.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Justin J. Botterill
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
11
|
Frigerio F, Pasqualini G, Craparotta I, Marchini S, van Vliet EA, Foerch P, Vandenplas C, Leclercq K, Aronica E, Porcu L, Pistorius K, Colas RA, Hansen TV, Perretti M, Kaminski RM, Dalli J, Vezzani A. n-3 Docosapentaenoic acid-derived protectin D1 promotes resolution of neuroinflammation and arrests epileptogenesis. Brain 2019; 141:3130-3143. [PMID: 30307467 PMCID: PMC6202571 DOI: 10.1093/brain/awy247] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/08/2018] [Indexed: 02/03/2023] Open
Abstract
Epilepsy therapy is based on drugs that treat the symptoms rather than the underlying mechanisms of the disease (epileptogenesis). There are no treatments for preventing seizures or improving disease prognosis, including neurological comorbidities. The search of pathogenic mechanisms of epileptogenesis highlighted that neuroinflammatory cytokines [i.e. interleukin-1β (IL-1β), tumour necrosis factor-α (Tnf-α)] are induced in human and experimental epilepsies, and contribute to seizure generation in animal models. A major role in controlling the inflammatory response is played by specialized pro-resolving lipid mediators acting on specific G-protein coupled receptors. Of note, the role that these pathways have in epileptogenic tissue remains largely unexplored. Using a murine model of epilepsy, we show that specialized pro-resolving mechanisms are activated by status epilepticus before the onset of spontaneous seizures, but with a marked delay as compared to the neuroinflammatory response. This was assessed by measuring the time course of mRNA levels of 5-lipoxygenase (Alox5) and 15-lipoxygenase (Alox15), the key biosynthetic enzymes of pro-resolving lipid mediators, versus Il1b and Tnfa transcripts and proteins. In the same hippocampal tissue, we found a similar delayed expression of two main pro-resolving receptors, the lipoxin A4 receptor/formyl peptide receptor 2 and the chemerin receptor. These receptors were also induced in the human hippocampus after status epilepticus and in patients with temporal lobe epilepsy. This evidence supports the hypothesis that the neuroinflammatory response is sustained by a failure to engage pro-resolving mechanisms during epileptogenesis. Lipidomic LC-MS/MS analysis showed that lipid mediator levels apt to resolve the neuroinflammatory response were also significantly altered in the hippocampus during epileptogenesis with a shift in the biosynthesis of several pro-resolving mediator families including the n-3 docosapentaenoic acid (DPA)-derived protectin D1. Of note, intracerebroventricular injection of this mediator during epileptogenesis in mice dose-dependently reduced the hippocampal expression of both Il1b and Tnfa mRNAs. This effect was associated with marked improvement in mouse weight recovery and rescue of cognitive deficit in the novel object recognition test. Notably, the frequency of spontaneous seizures was drastically reduced by 2-fold on average and the average seizure duration was shortened by 40% after treatment discontinuation. As a result, the total time spent in seizures was reduced by 3-fold in mice treated with n-3 DPA-derived protectin D1. Taken together, the present findings demonstrate that epilepsy is characterized by an inadequate engagement of resolution pathways. Boosting endogenous resolution responses significantly improved disease outcomes, providing novel treatment avenues.
Collapse
Affiliation(s)
- Federica Frigerio
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
| | - Giulia Pasqualini
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
| | - Ilaria Craparotta
- Department of Oncology, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
| | - Sergio Marchini
- Department of Oncology, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Amsterdam, The Netherlands
| | - Luca Porcu
- Department of Oncology, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
| | - Kimberly Pistorius
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Romain A Colas
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Trond V Hansen
- School of Pharmacy, Department of Pharmaceutical Chemistry, University of Oslo, Oslo, Norway
| | - Mauro Perretti
- William Harvey Research Institute, Queen Mary University of London, London, UK
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | | | - Jesmond Dalli
- William Harvey Research Institute, Queen Mary University of London, London, UK
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
- Correspondence regarding lipid mediators to: Jesmond Dalli, PhD Centre for inflammation and Therapeutic Innovation Queen Mary University of London Charterhouse Square, London, EC1M 6BQ, UK E-mail:
| | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCSS, Milano, Italy
- Correspondence regarding epileptogenesis to: Annamaria Vezzani, PhD Department of Neuroscience Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via G. La Masa 19, 20156 Milano, Italy E-mail:
| |
Collapse
|
12
|
Altered Synaptic Drive onto Birthdated Dentate Granule Cells in Experimental Temporal Lobe Epilepsy. J Neurosci 2019; 39:7604-7614. [PMID: 31270158 DOI: 10.1523/jneurosci.0654-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 12/29/2022] Open
Abstract
Dysregulated adult hippocampal neurogenesis occurs in many temporal lobe epilepsy (TLE) models. Most dentate granule cells (DGCs) generated in response to an epileptic insult develop features that promote increased excitability, including ectopic location, persistent hilar basal dendrites (HBDs), and mossy fiber sprouting. However, some appear to integrate normally and even exhibit reduced excitability compared to other DGCs. To examine the relationship between DGC birthdate, morphology, and network integration in a model of TLE, we retrovirally birthdated either early-born [EB; postnatal day (P)7] or adult-born (AB; P60) DGCs. Male rats underwent pilocarpine-induced status epilepticus (SE) or sham treatment at P56. Three to six months after SE or sham treatment, we used whole-cell patch-clamp and fluorescence microscopy to record spontaneous excitatory and inhibitory currents from birthdated DGCs. We found that both AB and EB populations of DGCs recorded from epileptic rats received increased excitatory input compared with age-matched controls. Interestingly, when AB populations were separated into normally integrated (normotopic) and aberrant (ectopic or HBD-containing) subpopulations, only the aberrant populations exhibited a relative increase in excitatory input (amplitude, frequency, and charge transfer). The ratio of excitatory-to-inhibitory input was most dramatically upregulated for ectopically localized DGCs. These data provide definitive physiological evidence that aberrant integration of post-SE, AB DGCs contributes to increased synaptic drive and support the idea that ectopic DGCs serve as putative hub cells to promote seizures.SIGNIFICANCE STATEMENT Adult dentate granule cell (DGC) neurogenesis is altered in rodent models of temporal lobe epilepsy (TLE). Some of the new neurons show abnormal morphology and integration, but whether adult-generated DGCs contribute to the development of epilepsy is controversial. We examined the synaptic inputs of age-defined populations of DGCs using electrophysiological recordings and fluorescent retroviral reporter birthdating. DGCs generated neonatally were compared with those generated in adulthood, and adult-born (AB) neurons with normal versus aberrant morphology or integration were examined. We found that AB, ectopically located DGCs exhibit the most pro-excitatory physiological changes, implicating this population in seizure generation or progression.
Collapse
|
13
|
Postnatal Increases in Axonal Conduction Velocity of an Identified Drosophila Interneuron Require Fast Sodium, L-Type Calcium and Shaker Potassium Channels. eNeuro 2019; 6:ENEURO.0181-19.2019. [PMID: 31253715 PMCID: PMC6709211 DOI: 10.1523/eneuro.0181-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 11/21/2022] Open
Abstract
During early postnatal life, speed up of signal propagation through many central and peripheral neurons has been associated with an increase in axon diameter or/and myelination. Especially in unmyelinated axons postnatal adjustments of axonal membrane conductances is potentially a third mechanism but solid evidence is lacking. Here, we show that axonal action potential (AP) conduction velocity in the Drosophila giant fiber (GF) interneuron, which is required for fast long-distance signal conduction through the escape circuit, is increased by 80% during the first day of adult life. Genetic manipulations indicate that this postnatal increase in AP conduction velocity in the unmyelinated GF axon is likely owed to adjustments of ion channel expression or properties rather than axon diameter increases. Specifically, targeted RNAi knock-down of either Para fast voltage-gated sodium, Shaker potassium (Kv1 homologue), or surprisingly, L-type like calcium channels counteracts postnatal increases in GF axonal conduction velocity. By contrast, the calcium-dependent potassium channel Slowpoke (BK) is not essential for postnatal speeding, although it also significantly increases conduction velocity. Therefore, we identified multiple ion channels that function to support fast axonal AP conduction velocity, but only a subset of these are regulated during early postnatal life to maximize conduction velocity. Despite its large diameter (∼7 µm) and postnatal regulation of multiple ionic conductances, mature GF axonal conduction velocity is still 20-60 times slower than that of vertebrate Aβ sensory axons and α motoneurons, thus unraveling the limits of long-range information transfer speed through invertebrate circuits.
Collapse
|
14
|
Goubert E, Altvater M, Rovira MN, Khalilov I, Mazzarino M, Sebastiani A, Schaefer MKE, Rivera C, Pellegrino C. Bumetanide Prevents Brain Trauma-Induced Depressive-Like Behavior. Front Mol Neurosci 2019; 12:12. [PMID: 30804751 PMCID: PMC6370740 DOI: 10.3389/fnmol.2019.00012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/14/2019] [Indexed: 01/24/2023] Open
Abstract
Brain trauma triggers a cascade of deleterious events leading to enhanced incidence of drug resistant epilepsies, depression, and cognitive dysfunctions. The underlying mechanisms leading to these alterations are poorly understood and treatment that attenuates those sequels are not available. Using controlled-cortical impact as an experimental model of brain trauma in adult mice, we found a strong suppressive effect of the sodium-potassium-chloride importer (NKCC1) specific antagonist bumetanide on the appearance of depressive-like behavior. We demonstrate that this alteration in behavior is associated with an impairment of post-traumatic secondary neurogenesis within the dentate gyrus of the hippocampus. The mechanism mediating the effect of bumetanide involves early transient changes in the expression of chloride regulatory proteins and qualitative changes in GABA(A) mediated transmission from hyperpolarizing to depolarizing after brain trauma. This work opens new perspectives in the early treatment of human post-traumatic induced depression. Our results strongly suggest that bumetanide might constitute an efficient prophylactic treatment to reduce neurological and psychiatric consequences of brain trauma.
Collapse
Affiliation(s)
- Emmanuelle Goubert
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Marc Altvater
- Department of Anesthesiology and Research Center Translational Neurosciences, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marie-Noelle Rovira
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Ilgam Khalilov
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France.,Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Morgane Mazzarino
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Anne Sebastiani
- Department of Anesthesiology and Research Center Translational Neurosciences, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael K E Schaefer
- Department of Anesthesiology and Research Center Translational Neurosciences, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudio Rivera
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Christophe Pellegrino
- INSERM, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| |
Collapse
|
15
|
Wixey JA, Chand KK, Pham L, Colditz PB, Bjorkman ST. Therapeutic potential to reduce brain injury in growth restricted newborns. J Physiol 2018; 596:5675-5686. [PMID: 29700828 DOI: 10.1113/jp275428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Brain injury in intrauterine growth restricted (IUGR) infants is a major contributing factor to morbidity and mortality worldwide. Adverse outcomes range from mild learning difficulties, to attention difficulties, neurobehavioral issues, cerebral palsy, epilepsy, and other cognitive and psychiatric disorders. While the use of medication to ameliorate neurological deficits in IUGR neonates has been identified as warranting urgent research for several years, few trials have been reported. This review summarises clinical trials focusing on brain protection in the IUGR newborn as well as therapeutic interventions trialled in animal models of IUGR. Therapeutically targeting mechanisms of brain injury in the IUGR neonate is fundamental to improving long-term neurodevelopmental outcomes. Inflammation is a key mechanism in neonatal brain injury; and therefore an appealing target. Ibuprofen, an anti-inflammatory drug currently used in the preterm neonate, may be a potential therapeutic candidate to treat brain injury in the IUGR neonate. To better understand the potential of ibuprofen and other therapeutic agents to be neuroprotective in the IUGR neonate, long-term follow-up information of neurodevelopmental outcomes must be studied. Where agents such as ibuprofen are shown to be effective, have a good safety profile and are relatively inexpensive, they can be widely adopted and lead to improved outcomes.
Collapse
Affiliation(s)
- Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Lily Pham
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - S Tracey Bjorkman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| |
Collapse
|
16
|
Scharfman HE, Kanner AM, Friedman A, Blümcke I, Crocker CE, Cendes F, Diaz-Arrastia R, Förstl H, Fenton AA, Grace AA, Palop J, Morrison J, Nehlig A, Prasad A, Wilcox KS, Jette N, Pohlmann-Eden B. Epilepsy as a Network Disorder (2): What can we learn from other network disorders such as dementia and schizophrenia, and what are the implications for translational research? Epilepsy Behav 2018; 78:302-312. [PMID: 29097123 PMCID: PMC5756681 DOI: 10.1016/j.yebeh.2017.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/18/2022]
Abstract
There is common agreement that many disorders of the central nervous system are 'complex', that is, there are many potential factors that influence the development of the disease, underlying mechanisms, and successful treatment. Most of these disorders, unfortunately, have no cure at the present time, and therapeutic strategies often have debilitating side effects. Interestingly, some of the 'complexities' of one disorder are found in another, and the similarities are often network defects. It seems likely that more discussions of these commonalities could advance our understanding and, therefore, have clinical implications or translational impact. With this in mind, the Fourth International Halifax Epilepsy Conference and Retreat was held as described in the prior paper, and this companion paper focuses on the second half of the meeting. Leaders in various subspecialties of epilepsy research were asked to address aging and dementia or psychosis in people with epilepsy (PWE). Commonalities between autism, depression, aging and dementia, psychosis, and epilepsy were the focus of the presentations and discussion. In the last session, additional experts commented on new conceptualization of translational epilepsy research efforts. Here, the presentations are reviewed, and salient points are highlighted.
Collapse
Affiliation(s)
- Helen E Scharfman
- Departments of Psychiatry, Neurosciences and Physiology, and the Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Andres M Kanner
- University of Miami, Miller School of Medicine, 1120 NW 14th Street, Room #1324, Miami, FL 33136, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ingmar Blümcke
- Neuropathological Institute, University Hospitals Erlangen, Germany
| | - Candice E Crocker
- Nova Scotia Early Psychosis Program, Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Fernando Cendes
- Department of Neurology, University of Campinas, 13083-888 Campinas, Sao Paulo, Brazil
| | - Ramon Diaz-Arrastia
- Centre for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, 12725 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Hans Förstl
- Department of Psychiatry, University of Munich, Klinikum rechts der Isar, Ismaninger Strabe 22, D-81675 Munich, Germany
| | - André A Fenton
- Centre for Neural Science, New York University, 4 Washington Place, Room 809, New York, NY 10003, USA
| | - Anthony A Grace
- University of Pittsburgh, 456 Langley Hall, 4200 Fifth Avenue, Pittsburgh, PA 15269, USA
| | - Jorge Palop
- Department of Neurology, Gladstone Institute, 1650 Owens Street, San Francisco, CA 94158-2261, USA
| | - Jason Morrison
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Astrid Nehlig
- INSERM U 1129, Hôpital Necker, Paris, Faculty of Medicine, Strasbourg, France
| | - Asuri Prasad
- Department of Pediatrics, Children's Hospital of Western Ontario, London, ON, Canada
| | - Karen S Wilcox
- Department of Pharmacology & Toxicology, Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT, USA
| | - Nathalie Jette
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Bernd Pohlmann-Eden
- Brain Repair Center, Life Science Research Institute, Dalhousie University, Room 229, PO Box 15000, Halifax, NS B3H4R2, Canada.
| |
Collapse
|
17
|
Repositioning of Somatic Golgi Apparatus Is Essential for the Dendritic Establishment of Adult-Born Hippocampal Neurons. J Neurosci 2017; 38:631-647. [PMID: 29217690 DOI: 10.1523/jneurosci.1217-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/02/2017] [Accepted: 10/29/2017] [Indexed: 01/22/2023] Open
Abstract
New dentate granule cells (DGCs) are continuously generated, and integrate into the preexisting hippocampal network in the adult brain. How an adult-born neuron with initially simple spindle-like morphology develops into a DGC, consisting of a single apical dendrite with further branches, remains largely unknown. Here, using retroviruses to birth date and manipulate newborn neurons, we examined initial dendritic formation and possible underlying mechanisms. We found that GFP-expressing newborn cells began to establish a DGC-like morphology at ∼7 d after birth, with a primary dendrite pointing to the molecular layer, but at this stage, with several neurites in the neurogenic zone. Interestingly, the Golgi apparatus, an essential organelle for neurite growth and maintenance, was dynamically repositioning in the soma of newborn cells during this initial integration stage. Two weeks after birth, by which time most neurites in the neurogenic zone were eliminated, a compact Golgi apparatus was positioned exclusively at the base of the primary dendrite. We analyzed the presence of Golgi-associated genes using single-cell transcriptomes of newborn DGCs, and among Golgi-related genes, found the presence of STK25 and STRAD, regulators of embryonic neuronal development. When we knocked down either of these two proteins, we found Golgi mislocalization and extensive aberrant dendrite formation. Furthermore, overexpression of a mutated form of STRAD, underlying the disorder polyhydramnios, megalencephaly, and symptomatic epilepsy, characterized by abnormal brain development and intractable epilepsy, caused similar defects in Golgi localization and dendrite formation in adult-born neurons. Together, our findings reveal a role for Golgi repositioning in regulating the initial integration of adult-born DGCs.SIGNIFICANCE STATEMENT Since the discovery of the continuous generation of new neurons in the adult hippocampus, extensive effort was directed toward understanding the functional contribution of these newborn neurons to the existing hippocampal circuit and associated behaviors, while the molecular mechanisms controlling their early morphological integration are less well understood. Dentate granule cells (DGCs) have a single, complex, apical dendrite. The events leading adult-born DGCs' to transition from simple spindle-like morphology to mature dendrite morphology are largely unknown. We studied establishment of newborn DGCs dendritic pattern and found it was mediated by a signaling pathway regulating precise localization of the Golgi apparatus. Furthermore, this Golgi-associated mechanism for dendrite establishment might be impaired in a human genetic epilepsy syndrome, polyhydramnios, megalencephaly, and symptomatic epilepsy.
Collapse
|
18
|
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
|
19
|
Bermudez-Hernandez K, Lu YL, Moretto J, Jain S, LaFrancois JJ, Duffy AM, Scharfman HE. Hilar granule cells of the mouse dentate gyrus: effects of age, septotemporal location, strain, and selective deletion of the proapoptotic gene BAX. Brain Struct Funct 2017; 222:3147-3161. [PMID: 28314928 PMCID: PMC5601016 DOI: 10.1007/s00429-017-1391-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 02/20/2017] [Indexed: 12/11/2022]
Abstract
The dentate gyrus (DG) principal cells are glutamatergic granule cells (GCs), and they are located in a compact cell layer. However, GCs are also present in the adjacent hilar region, but have been described in only a few studies. Therefore, we used the transcription factor prospero homeobox 1 (Prox1) to quantify GCs at postnatal day (PND) 16, 30, and 60 in a common mouse strain, C57BL/6J mice. At PND16, there was a large population of Prox1-immunoreactive (ir) hilar cells, with more in the septal than temporal hippocampus. At PND30 and 60, the size of the hilar Prox1-ir cell population was reduced. Similar numbers of hilar Prox1-expressing cells were observed in PND30 and 60 Swiss Webster mice. Prox1 is usually considered to be a marker of postmitotic GCs. However, many Prox1-ir hilar cells, especially at PND16, were not double-labeled with NeuN, a marker typically found in mature neurons. Most hilar Prox1-positive cells at PND16 co-expressed doublecortin (DCX) and calretinin, markers of immature GCs. Double-labeling with a marker of actively dividing cells, Ki67, was not detected. These results suggest that, surprisingly, a large population of cells in the hilus at PND16 are immature GCs (Type 2b and Type 3 cells). We also asked whether hilar Prox1-ir cell numbers are modifiable. To examine this issue, we conditionally deleted the proapoptotic gene BAX in Nestin-expressing cells at a time when there are numerous immature GCs in the hilus, PND2-8. When these mice were examined at PND60, the numbers of Prox1-ir hilar cells were significantly increased compared to control mice. However, deletion of BAX did not appear to change the proportion that co-expressed NeuN, suggesting that the size of the hilar Prox1-expressing population is modifiable. However, deleting BAX, a major developmental disruption, does not appear to change the proportion that ultimately becomes neurons.
Collapse
Affiliation(s)
- Keria Bermudez-Hernandez
- New York University Langone Medical Center, 227 East 30th Street, 7th Floor, One Park Avenue, New York, NY, 10016, USA.
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA.
| | - Yi-Ling Lu
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Jillian Moretto
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Swati Jain
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - John J LaFrancois
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Aine M Duffy
- New York University Langone Medical Center, 227 East 30th Street, 7th Floor, One Park Avenue, New York, NY, 10016, USA
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Helen E Scharfman
- New York University Langone Medical Center, 227 East 30th Street, 7th Floor, One Park Avenue, New York, NY, 10016, USA
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| |
Collapse
|
20
|
Long-term effects of enriched environment following neonatal hypoxia-ischemia on behavior, BDNF and synaptophysin levels in rat hippocampus: Effect of combined treatment with G-CSF. Brain Res 2017; 1667:55-67. [DOI: 10.1016/j.brainres.2017.05.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 04/13/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022]
|
21
|
Brulet R, Zhu J, Aktar M, Hsieh J, Cho KO. Mice with conditional NeuroD1 knockout display reduced aberrant hippocampal neurogenesis but no change in epileptic seizures. Exp Neurol 2017; 293:190-198. [PMID: 28427858 PMCID: PMC5503142 DOI: 10.1016/j.expneurol.2017.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 12/19/2022]
Abstract
Adult neurogenesis is significantly increased in the hippocampus of rodent models of temporal lobe epilepsy (TLE). These adult-generated neurons have recently been shown to play a contributing role in the development of spontaneous recurrent seizures (SRS). In order to eventually target pro-epileptic adult neurogenesis in the clinical setting, it will be important to identify molecular players involved in the control of aberrant neurogenesis after seizures. Here, we focused on NeuroD1 (ND1), a member of the bHLH family of transcription factors previously shown to play an essential role in the differentiation and maturation of adult-generated neurons in the hippocampus. Wild-type mice treated with pilocarpine to induce status epilepticus (SE) showed a significant up-regulation of NeuroD1+ immature neuroblasts located in both the granule cell layer (GCL), and ectopically localized to the hilar region of the hippocampus. As expected, conditional knockout (cKO) of NeuroD1 in Nestin-expressing stem/progenitors and their progeny led to a reduction in the number of NeuroD1+ adult-generated neurons after pilocarpine treatment compared to WT littermates. Surprisingly, there was no change in SRS in NeuroD1 cKO mice, suggesting that NeuroD1 cKO fails to reduce aberrant neurogenesis below the threshold needed to impact SRS. Consistent with this conclusion, the total number of adult-generated neurons in the pilocarpine model, especially the total number of Prox1+ hilar ectopic granule cells were unchanged after NeuroD1 cKO, suggesting strategies to reduce SRS will need to achieve a greater removal of aberrant adult-generated neurons.
Collapse
Affiliation(s)
- Rebecca Brulet
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jingfei Zhu
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mahafuza Aktar
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenny Hsieh
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Kyung-Ok Cho
- Department of Pharmacology, Catholic Neuroscience Institute, School of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
22
|
Zhou Z, Liu T, Sun X, Mu X, Zhu G, Xiao T, Zhao M, Zhao C. CXCR4 antagonist AMD3100 reverses the neurogenesis promoted by enriched environment and suppresses long-term seizure activity in adult rats of temporal lobe epilepsy. Behav Brain Res 2017; 322:83-91. [DOI: 10.1016/j.bbr.2017.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/02/2017] [Accepted: 01/06/2017] [Indexed: 12/16/2022]
|
23
|
Kourdougli N, Pellegrino C, Renko JM, Khirug S, Chazal G, Kukko-Lukjanov TK, Lauri SE, Gaiarsa JL, Zhou L, Peret A, Castrén E, Tuominen RK, Crépel V, Rivera C. Depolarizing γ-aminobutyric acid contributes to glutamatergic network rewiring in epilepsy. Ann Neurol 2017; 81:251-265. [PMID: 28074534 DOI: 10.1002/ana.24870] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 12/16/2023]
Abstract
OBJECTIVE Rewiring of excitatory glutamatergic neuronal circuits is a major abnormality in epilepsy. Besides the rewiring of excitatory circuits, an abnormal depolarizing γ-aminobutyric acidergic (GABAergic) drive has been hypothesized to participate in the epileptogenic processes. However, a remaining clinically relevant question is whether early post-status epilepticus (SE) evoked chloride dysregulation is important for the remodeling of aberrant glutamatergic neuronal circuits. METHODS Osmotic minipumps were used to infuse intracerebrally a specific inhibitor of depolarizing GABAergic transmission as well as a functionally blocking antibody toward the pan-neurotrophin receptor p75 (p75NTR ). The compounds were infused between 2 and 5 days after pilocarpine-induced SE. Immunohistochemistry for NKCC1, KCC2, and ectopic recurrent mossy fiber (rMF) sprouting as well as telemetric electroencephalographic and electrophysiological recordings were performed at day 5 and 2 months post-SE. RESULTS Blockade of NKCC1 after SE with the specific inhibitor bumetanide restored NKCC1 and KCC2 expression, normalized chloride homeostasis, and significantly reduced the glutamatergic rMF sprouting within the dentate gyrus. This mechanism partially involves p75NTR signaling, as bumetanide application reduced SE-induced p75NTR expression and functional blockade of p75NTR decreased rMF sprouting. The early transient (3 days) post-SE infusion of bumetanide reduced rMF sprouting and recurrent seizures in the chronic epileptic phase. INTERPRETATION Our findings show that early post-SE abnormal depolarizing GABA and p75NTR signaling fosters a long-lasting rearrangement of glutamatergic network that contributes to the epileptogenic process. This finding defines promising and novel targets to constrain reactive glutamatergic network rewiring in adult epilepsy. Ann Neurol 2017;81:251-265.
Collapse
Affiliation(s)
- Nazim Kourdougli
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | - Christophe Pellegrino
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | - Juho-Matti Renko
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | | | - Geneviève Chazal
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | | | - Sari E Lauri
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jean-Luc Gaiarsa
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | - Liang Zhou
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Angélique Peret
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Valérie Crépel
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
| | - Claudio Rivera
- Inserm Unit 901, Inmed, Marseille, France
- Mixed Unit of Research S901, Aix-Marseille University, Marseille, France
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Taylor SR, Smith CM, Keeley KL, McGuone D, Dodge CP, Duhaime AC, Costine BA. Neuroblast Distribution after Cortical Impact Is Influenced by White Matter Injury in the Immature Gyrencephalic Brain. Front Neurosci 2016; 10:387. [PMID: 27601978 PMCID: PMC4994423 DOI: 10.3389/fnins.2016.00387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/08/2016] [Indexed: 11/13/2022] Open
Abstract
Cortical contusions are a common type of traumatic brain injury (TBI) in children. Current knowledge of neuroblast response to cortical injury arises primarily from studies utilizing aspiration or cryoinjury in rodents. In infants and children, cortical impact affects both gray and white matter and any neurogenic response may be complicated by the large expanse of white matter between the subventricular zone (SVZ) and the cortex, and the large number of neuroblasts in transit along the major white matter tracts to populate brain regions. Previously, we described an age-dependent increase of neuroblasts in the SVZ in response to cortical impact in the immature gyrencephalic brain. Here, we investigate if neuroblasts target the injury, if white matter injury influences repair efforts, and if postnatal population of brain regions are disrupted. Piglets received a cortical impact to the rostral gyrus cortex or sham surgery at postnatal day (PND) 7, BrdU 2 days prior to (PND 5 and 6) or after injury (PND 7 and 8), and brains were collected at PND 14. Injury did not alter the number of neuroblasts in the white matter between the SVZ and the rostral gyrus. In the gray matter of the injury site, neuroblast density was increased in cavitated lesions, and the number of BrdU(+) neuroblasts was increased, but comprised less than 1% of all neuroblasts. In the white matter of the injury site, neuroblasts with differentiating morphology were densely arranged along the cavity edge. In a ventral migratory stream, neuroblast density was greater in subjects with a cavitated lesion, indicating that TBI may alter postnatal development of regions supplied by that stream. Cortical impact in the immature gyrencephalic brain produced complicated and variable lesions, increased neuroblast density in cavitated gray matter, resulted in potentially differentiating neuroblasts in the white matter, and may alter the postnatal population of brain regions utilizing a population of neuroblasts that were born prior to PND 5. This platform may be useful to continue to study potential complications of white matter injury and alterations of postnatal population of brain regions, which may contribute to the chronic effects of TBI in children.
Collapse
Affiliation(s)
- Sabrina R Taylor
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Charlestown, MA, USA
| | - Colin M Smith
- Brain Trauma Lab, Department of Neurosurgery, Massachusetts General Hospital Boston, MA, USA
| | - Kristen L Keeley
- Brain Trauma Lab, Department of Neurosurgery, Massachusetts General Hospital Boston, MA, USA
| | | | - Carter P Dodge
- Department of Anesthesiology, Dartmouth Medical School, Children's Hospital at Dartmouth Lebanon, PA, USA
| | - Ann-Christine Duhaime
- Brain Trauma Lab, Department of Neurosurgery, Massachusetts General HospitalBoston, MA, USA; Department of Neurosurgery, Harvard Medical SchoolBoston, MA, USA
| | - Beth A Costine
- Brain Trauma Lab, Department of Neurosurgery, Massachusetts General HospitalBoston, MA, USA; Department of Neurosurgery, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Schouten M, Fratantoni SA, Hubens CJ, Piersma SR, Pham TV, Bielefeld P, Voskuyl RA, Lucassen PJ, Jimenez CR, Fitzsimons CP. MicroRNA-124 and -137 cooperativity controls caspase-3 activity through BCL2L13 in hippocampal neural stem cells. Sci Rep 2015. [PMID: 26207921 PMCID: PMC4513647 DOI: 10.1038/srep12448] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Adult neurogenesis continuously contributes new neurons to hippocampal circuits and the programmed death of a subset of immature cells provides a primary mechanism controlling this contribution. Epileptic seizures induce strong structural changes in the hippocampus, including the induction of adult neurogenesis, changes in gene expression and mitochondrial dysfunction, which may all contribute to epileptogenesis. However, a possible interplay between this factors remains largely unexplored. Here, we investigated gene expression changes in the hippocampal dentate gyrus shortly after prolonged seizures induced by kainic acid, focusing on mitochondrial functions. Using comparative proteomics, we identified networks of proteins differentially expressed shortly after seizure induction, including members of the BCL2 family and other mitochondrial proteins. Within these networks, we report for the first time that the atypical BCL2 protein BCL2L13 controls caspase-3 activity and cytochrome C release in neural stem/progenitor cells. Furthermore, we identify BCL2L13 as a novel target of the cooperative action of microRNA-124 and microRNA-137, both upregulated shortly after seizure induction. This cooperative microRNA-mediated fine-tuning of BCL2L13 expression controls casp3 activity, favoring non-apoptotic caspase-3 functions in NSPC exposed to KA and thereby may contribute to the early neurogenic response to epileptic seizures in the dentate gyrus.
Collapse
Affiliation(s)
- Marijn Schouten
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH, Amsterdam, The Netherlands
| | - Silvina A Fratantoni
- Oncoproteomics Laboratory, Cancer Center, Free University Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Chantal J Hubens
- 1] Division of Pharmacology, LACDR, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands [2] Foundation of Epilepsy Institutes of The Netherlands (SEIN), Achterweg 5, 2103 SW, Heemstede, The Netherlands
| | - Sander R Piersma
- Oncoproteomics Laboratory, Cancer Center, Free University Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Thang V Pham
- Oncoproteomics Laboratory, Cancer Center, Free University Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Pascal Bielefeld
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH, Amsterdam, The Netherlands
| | - Rob A Voskuyl
- 1] Division of Pharmacology, LACDR, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands [2] Foundation of Epilepsy Institutes of The Netherlands (SEIN), Achterweg 5, 2103 SW, Heemstede, The Netherlands
| | - Paul J Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH, Amsterdam, The Netherlands
| | - Connie R Jimenez
- Oncoproteomics Laboratory, Cancer Center, Free University Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Carlos P Fitzsimons
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH, Amsterdam, The Netherlands
| |
Collapse
|
28
|
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
|
29
|
Dunleavy M, Schindler CK, Shinoda S, Crilly S, Henshall DC. Neurogenic function in rats with unilateral hippocampal sclerosis that experienced early-life status epilepticus. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2014; 6:199-208. [PMID: 25755841 PMCID: PMC4348706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/13/2014] [Indexed: 06/04/2023]
Abstract
Status epilepticus in the adult brain invariably causes an increase in hippocampal neurogenesis and the appearance of ectopic cells and this has been implicated as a causal factor in epileptogenesis. The effect of status epilepticus on neurogenesis in the developing brain is less well characterized and models of early-life seizures typically do not reproduce the hippocampal damage common to human mesial temporal sclerosis. We recently reported that evoking status epilepticus by intra-amygdala microinjection of kainic acid in post-natal (P) day 10 rats caused substantial acute neuronal death within the ipsilateral hippocampus and rats later developed unilateral hippocampal sclerosis and spontaneous recurrent seizures. Here, we examined the expression of a selection of genes associated with neurogenesis and assessed neurogenic function in this model. Protein levels of several markers of neurogenesis including polysialic acid neural cell adhesion molecule, neuroD and doublecortin were reduced in the hippocampus three days after status epilepticus in P10 rats. In contrast, protein levels of neurogenesis markers were similar to control in rats at P55. Pulse-chase experiments using thymidine analogues suggested there was a reduction in new neurons at 72 h after status epilepticus in P10 rats, whereas numbers of new neurons labelled in epileptic rats at P55 with hippocampal sclerosis were similar to controls. The present study suggests that status epilepticus in the immature brain suppresses neurogenesis but the neurogenic potential is retained in animals that later develop hippocampal sclerosis.
Collapse
Affiliation(s)
- Mark Dunleavy
- Department of Physiology & Medical Physics, Royal College of Surgeons in IrelandDublin, Ireland
| | - Clara K Schindler
- Robert S. Dow Neurobiology Laboratories, Legacy ResearchPortland, OR, USA
| | - Sachiko Shinoda
- Robert S. Dow Neurobiology Laboratories, Legacy ResearchPortland, OR, USA
- Department of Neurosurgery, Mie University School of MedicineMie, Tsu, Japan
| | - Shane Crilly
- Department of Physiology & Medical Physics, Royal College of Surgeons in IrelandDublin, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in IrelandDublin, Ireland
- Irish Centre for Fetal and Neonatal Translational ResearchCork, Ireland
| |
Collapse
|
30
|
Kight KE, McCarthy MM. Using sex differences in the developing brain to identify nodes of influence for seizure susceptibility and epileptogenesis. Neurobiol Dis 2014; 72 Pt B:136-43. [PMID: 24892888 PMCID: PMC5322568 DOI: 10.1016/j.nbd.2014.05.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 12/12/2022] Open
Abstract
Sexual differentiation of the developing brain organizes the neural architecture differently between males and females, and the main influence on this process is exposure to gonadal steroids during sensitive periods of prenatal and early postnatal development. Many molecular and cellular processes are influenced by steroid hormones in the developing brain, including gene expression, cell birth and death, neurite outgrowth and synaptogenesis, and synaptic activity. Perturbations in these processes can alter neuronal excitability and circuit activity, leading to increased seizure susceptibility and the promotion of pathological processes that constitute epileptogenesis. In this review, we will provide a general overview of sex differences in the early developing brain that may be relevant for altered seizure susceptibility in early life, focusing on limbic areas of the brain. Sex differences that have the potential to alter the progress of epileptogenesis are evident at molecular and cellular levels in the developing brain, and include differences in neuronal excitability, response to environmental insult, and epigenetic control of gene expression. Knowing how these processes differ between the sexes can help us understand fundamental mechanisms underlying gender differences in seizure susceptibility and epileptogenesis.
Collapse
Affiliation(s)
- Katherine E Kight
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Margaret M McCarthy
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
31
|
Shetty AK. Hippocampal injury-induced cognitive and mood dysfunction, altered neurogenesis, and epilepsy: can early neural stem cell grafting intervention provide protection? Epilepsy Behav 2014; 38:117-24. [PMID: 24433836 PMCID: PMC4742318 DOI: 10.1016/j.yebeh.2013.12.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/02/2013] [Indexed: 01/25/2023]
Abstract
Damage to the hippocampus can occur through many causes including head trauma, ischemia, stroke, status epilepticus, and Alzheimer's disease. Certain changes such as increased levels of neurogenesis and elevated concentrations of multiple neurotrophic factors that ensue in the acute phase after injury seem beneficial for restraining hippocampal dysfunction. However, many alterations that arise in the intermediate to chronic phase after injury such as abnormal migration of newly born neurons, aberrant synaptic reorganization, progressive loss of inhibitory gamma-amino butyric acid positive interneurons including those expressing reelin, greatly declined neurogenesis, and sustained inflammation are detrimental. Consequently, the net effect of postinjury plasticity in the hippocampus remains inadequate for promoting significant functional recovery. Hence, ideal therapeutic interventions ought to be efficient for restraining these detrimental changes in order to block the propensity of most hippocampal injuries to evolve into learning deficits, memory dysfunction, depression, and temporal lobe epilepsy. Neural stem cell (NSC) grafting into the hippocampus early after injury appears alluring from this perspective because several recent studies have demonstrated the therapeutic value of this intervention, especially for preventing/easing memory dysfunction, depression, and temporal lobe epilepsy development in the chronic phase after injury. These beneficial effects of NSC grafting appeared to be mediated through considerable modulation of aberrant hippocampal postinjury plasticity with additions of new inhibitory gamma-amino butyric acid positive interneurons and astrocytes secreting a variety of neurotrophic factors and anticonvulsant proteins. This review presents advancements made in NSC grafting therapy for treating hippocampal injury in animal models of excitotoxic injury, traumatic brain injury, Alzheimer's disease, and status epilepticus; potential mechanisms of functional recovery mediated by NSC grafts placed early after hippocampal injury; and issues that need to be resolved prior to considering clinical application of NSC grafting for hippocampal injury.
Collapse
Affiliation(s)
- Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, TX, USA; Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA; Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA.
| |
Collapse
|
32
|
Botterill JJ, Guskjolen AJ, Marks WN, Caruncho HJ, Kalynchuk LE. Limbic but not non-limbic kindling impairs conditioned fear and promotes plasticity of NPY and its Y2 receptor. Brain Struct Funct 2014; 220:3641-55. [PMID: 25146309 DOI: 10.1007/s00429-014-0880-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/13/2014] [Indexed: 12/01/2022]
Abstract
Epileptic seizures negatively affect cognition. However, the mechanisms that contribute to cognitive impairments after seizures are largely unknown. Here, we examined the effects of long-term kindling (i.e., 99 stimulations) of limbic (basolateral amygdala, dorsal hippocampus) and non-limbic (caudate nucleus) brain sites on conditioned fear and hippocampal plasticity. We first showed that kindling had no effect on acquisition of a hippocampal-dependent trace fear-conditioning task but limbic kindling impaired the retrieval of these fear memories. To determine the relationship between memory and hippocampal neuronal activity, we examined the expression of Fos protein 90 min after memory retrieval (i.e., 4 days after the last kindling stimulation). We found that limbic kindling, but not non-limbic kindling, decreased Fos expression in the granule cell layer, hilus, CA3 pyramidal cell layer, and CA1 pyramidal cell layer. Next, to investigate a mechanism that could contribute to dampen hippocampal neuronal activity in limbic-kindled rats, we focused on the endogenous anticonvulsant neuropeptide Y (NPY), which is expressed in a subset of GABAergic interneurons and can prevent glutamate release through interactions with its Y2 receptor. We found that limbic kindling significantly decreased the number of NPY-immunoreactive cells in several hippocampal subfields despite minimal staining of the neurodegenerative marker Fluoro-Jade B. However, we also noted that limbic kindling enhanced NPY immunoreactivity throughout the mossy fiber pathway. In these same regions, we observed limbic kindling-induced de novo expression of the NPY Y2 receptor. These novel findings demonstrate the site-specific effects of kindling on cognition and NPY plasticity, and they provide evidence that altered hippocampal NPY after limbic seizures coincides with dampened neural activity and cognitive impairments.
Collapse
Affiliation(s)
- J J Botterill
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - A J Guskjolen
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - W N Marks
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - H J Caruncho
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - L E Kalynchuk
- Department of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
33
|
Lasarge CL, Danzer SC. Mechanisms regulating neuronal excitability and seizure development following mTOR pathway hyperactivation. Front Mol Neurosci 2014; 7:18. [PMID: 24672426 PMCID: PMC3953715 DOI: 10.3389/fnmol.2014.00018] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 02/27/2014] [Indexed: 01/19/2023] Open
Abstract
The phosphatidylinositol-3-kinase/phosphatase and tensin homolog (PTEN)-mammalian target of rapamycin (mTOR) pathway regulates a variety of neuronal functions, including cell proliferation, survival, growth, and plasticity. Dysregulation of the pathway is implicated in the development of both genetic and acquired epilepsies. Indeed, several causal mutations have been identified in patients with epilepsy, the most prominent of these being mutations in PTEN and tuberous sclerosis complexes 1 and 2 (TSC1, TSC2). These genes act as negative regulators of mTOR signaling, and mutations lead to hyperactivation of the pathway. Animal models deleting PTEN, TSC1, and TSC2 consistently produce epilepsy phenotypes, demonstrating that increased mTOR signaling can provoke neuronal hyperexcitability. Given the broad range of changes induced by altered mTOR signaling, however, the mechanisms underlying seizure development in these animals remain uncertain. In transgenic mice, cell populations with hyperactive mTOR have many structural abnormalities that support recurrent circuit formation, including somatic and dendritic hypertrophy, aberrant basal dendrites, and enlargement of axon tracts. At the functional level, mTOR hyperactivation is commonly, but not always, associated with enhanced synaptic transmission and plasticity. Moreover, these populations of abnormal neurons can affect the larger network, inducing secondary changes that may explain paradoxical findings reported between cell and network functioning in different models or at different developmental time points. Here, we review the animal literature examining the link between mTOR hyperactivation and epileptogenesis, emphasizing the impact of enhanced mTOR signaling on neuronal form and function.
Collapse
Affiliation(s)
- Candi L Lasarge
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - 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, University of Cincinnati Cincinnati, OH, USA
| |
Collapse
|
34
|
Saaltink DJ, Vreugdenhil E. Stress, glucocorticoid receptors, and adult neurogenesis: a balance between excitation and inhibition? Cell Mol Life Sci 2014; 71:2499-515. [PMID: 24522255 PMCID: PMC4055840 DOI: 10.1007/s00018-014-1568-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/26/2013] [Accepted: 01/16/2014] [Indexed: 02/06/2023]
Abstract
Adult neurogenesis, the birth of new neurons in the mature brain, has attracted considerable attention in the last decade. One of the earliest identified and most profound factors that affect adult neurogenesis both positively and negatively is stress. Here, we review the complex interplay between stress and adult neurogenesis. In particular, we review the role of the glucocorticoid receptor, the main mediator of the stress response in the proliferation, differentiation, migration, and functional integration of newborn neurons in the hippocampus. We review a multitude of mechanisms regulating glucocorticoid receptor activity in relationship to adult neurogenesis. We postulate a novel concept in which the level of glucocorticoid receptor expression directly regulates the excitation-inhibition balance, which is key for proper neurogenesis. We furthermore argue that an excitation-inhibition dis-balance may underlie aberrant functional integration of newborn neurons that is associated with psychiatric and paroxysmal brain disorders.
Collapse
Affiliation(s)
- Dirk-Jan Saaltink
- Department of Medical Pharmacology, Leiden University Medical Center/Leiden Amsterdam Center for Drug Research, 2300 RC, Leiden, The Netherlands
| | | |
Collapse
|
35
|
Scharfman HE, Brooks-Kayal AR. Is plasticity of GABAergic mechanisms relevant to epileptogenesis? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:133-50. [PMID: 25012373 DOI: 10.1007/978-94-017-8914-1_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Numerous changes in GABAergic neurons, receptors, and inhibitory mechanisms have been described in temporal lobe epilepsy (TLE), either in humans or in animal models. Nevertheless, there remains a common assumption that epilepsy can be explained by simply an insufficiency of GABAergic inhibition. Alternatively, investigators have suggested that there is hyperinhibition that masks an underlying hyperexcitability. Here we examine the status epilepticus (SE) models of TLE and focus on the dentate gyrus of the hippocampus, where a great deal of data have been collected. The types of GABAergic neurons and GABAA receptors are summarized under normal conditions and after SE. The role of GABA in development and in adult neurogenesis is discussed. We suggest that instead of "too little or too much" GABA there is a complexity of changes after SE that makes the emergence of chronic seizures (epileptogenesis) difficult to understand mechanistically, and difficult to treat. We also suggest that this complexity arises, at least in part, because of the remarkable plasticity of GABAergic neurons and GABAA receptors in response to insult or injury.
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA,
| | | |
Collapse
|
36
|
Caspase 3 involves in neuroplasticity, microglial activation and neurogenesis in the mice hippocampus after intracerebral injection of kainic acid. J Biomed Sci 2013; 20:90. [PMID: 24313976 PMCID: PMC4028745 DOI: 10.1186/1423-0127-20-90] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/02/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The roles of caspase 3 on the kainic acid-mediated neurodegeneration, dendritic plasticity alteration, neurogenesis, microglial activation and gliosis are not fully understood. Here, we investigate hippocampal changes using a mouse model that receive a single kainic acid-intracerebral ventricle injection. The effects of caspase 3 inhibition on these changes were detected during a period of 1 to 7 days post kainic acid injection. RESULT Neurodegeneration was assessed by Fluoro-Jade B staining and neuronal nuclei protein (NeuN) immunostaining. Neurogenesis, gliosis, neuritic plasticity alteration and caspase 3 activation were examined using immunohistochemistry. Dendritic plasticity, cleavvage-dependent activation of calcineurin A and glial fibrillary acidic protein cleavage were analyzed by immunoblotting. We found that kainic acid not only induced neurodegeneration but also arouse several caspase 3-mediated molecular and cellular changes including dendritic plasticity, neurogenesis, and gliosis. The acute caspase 3 activation occurred in pyramidal neurons as well as in hilar interneurons. The delayed caspase 3 activation occurred in astrocytes. The co-injection of caspase 3 inhibitor did not rescue kainic acid-mediated neurodegeneration but seriously and reversibly disturb the structural integrity of axon and dendrite. The kainic acid-induced events include microglia activation, the proliferation of radial glial cells, neurogenesis, and calcineurin A cleavage were significantly inhibited by the co-injection of caspase 3 inhibitor, suggesting the direct involvement of caspase 3 in these events. Alternatively, the kainic acid-mediated astrogliosis is not caspase 3-dependent, although caspase 3 cleavage of glial fibrillary acidic protein occurred. CONCLUSIONS Our results provide the first direct evidence of a causal role of caspase 3 activation in the cellular changes during kainic acid-mediated excitotoxicity. These findings may highlight novel pharmacological strategies to arrest disease progression and control seizures that are refractory to classical anticonvulsant treatment.
Collapse
|
37
|
Shetty AK. Prospects of levetiracetam as a neuroprotective drug against status epilepticus, traumatic brain injury, and stroke. Front Neurol 2013; 4:172. [PMID: 24204362 PMCID: PMC3816384 DOI: 10.3389/fneur.2013.00172] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/21/2013] [Indexed: 01/08/2023] Open
Abstract
Levetiracetam (LEV) is an anti-epileptic drug commonly used for the treatment of partial onset and generalized seizures. In addition to its neuromodulatory and neuroinhibitory effects via its binding to the synaptic vesicle protein SV2A, multiple studies have suggested neuroprotective properties for LEV in both epileptic and non-epileptic conditions. The purpose of this review is to discuss the extent of LEV-mediated protection seen in different neurological conditions, the potential of LEV for easing epileptogenesis, and the possible mechanisms that underlie the protective properties of LEV. LEV has been found to be particularly beneficial for restraining seizures in animal models of spontaneous epilepsy, acute seizures, and status epilepticus (SE). However, its ability for easing epileptogenesis and cognitive dysfunction following SE remains controversial with some studies implying favorable outcomes and others reporting no beneficial effects. Efficacy of LEV as a neuroprotective drug against traumatic brain injury (TBI) has received much attention. While animal studies in TBI models have showed significant neuroprotection and improvements in motor and memory performance with LEV treatment, clinical studies suggest that LEV has similar efficacy as phenytoin in terms of its ability to prevent post-traumatic epilepsy. LEV treatment for TBI is also reported to have fewer adverse effects and monitoring considerations but electroencephalographic recordings suggest the presence of increased seizure tendency. Studies on stroke imply that LEV is a useful alternative to carbamazepine for preventing post-stroke seizures in terms of efficacy and safety. Thus, LEV treatment has promise for restraining SE-, TBI-, or stroke-induced chronic epilepsy. Nevertheless, additional studies are needed to ascertain the most apt dose, timing of intervention, and duration of treatment after the initial precipitating injury and the mechanisms underlying LEV-mediated beneficial effects.
Collapse
Affiliation(s)
- Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White , Temple, TX , USA ; Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System , Temple, TX , USA ; Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine , College Station, TX , USA
| |
Collapse
|
38
|
Biagini G, Rustichelli C, Curia G, Vinet J, Lucchi C, Pugnaghi M, Meletti S. Neurosteroids and epileptogenesis. J Neuroendocrinol 2013; 25:980-90. [PMID: 23763517 DOI: 10.1111/jne.12063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/21/2013] [Accepted: 06/09/2013] [Indexed: 12/22/2022]
Abstract
Epileptogenesis is defined as the latent period at the end of which spontaneous recurrent seizures occur. This concept has been recently re-evaluated to include exacerbation of clinically-manifested epilepsy. Thus, in patients affected by pharmacoresistant seizures, the progression toward a worse condition may be viewed as the result of a durable epileptogenic process. However, the mechanism potentially responsible for this progression remains unclear. Neuroinflammation has been consistently detected both in the latent period and in the chronic phase of epilepsy, especially when brain damage is present. This phenomenon is accompanied by glial cell reaction, leading to gliosis. We have previously described rats presenting an increased expression of the cytochrome P450 cholesterol side-chain cleavage (P450scc) enzyme, during the latent period, in glial cells of the hippocampus. The P450scc enzyme is critically involved in the synthesis of neurosteroids and its up-regulation is associated with a delayed appearance of spontaneous recurrent seizures in rats that experienced status epilepticus induced by pilocarpine. Moreover, by decreasing the synthesis of neurosteroids able to promote inhibition, such as allopregnanolone, through the administration of the 5α-reductase blocker finasteride, it is possible to terminate the latent period in pilocarpine-treated rats. Finasteride was also found to promote seizures in the chronic period of epileptic rats, suggesting that neurosteroids are continuously produced to counteract seizures. In humans, exacerbation of epilepsy has been also described in patients occasionally exposed to finasteride. Overall, these findings suggest a major role of neurosteroids in the progression of epilepsy and a possible antiepileptogenic role of allopregnanolone and cognate molecules.
Collapse
Affiliation(s)
- G Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Lakhina V, Subramanian L, Huilgol D, Shetty AS, Vaidya VA, Tole S. Seizure evoked regulation of LIM-HD genes and co-factors in the postnatal and adult hippocampus. F1000Res 2013; 2:205. [PMID: 25110573 PMCID: PMC4111125 DOI: 10.12688/f1000research.2-205.v1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2013] [Indexed: 12/03/2022] Open
Abstract
The LIM-homeodomain (LIM-HD) family of transcription factors is well known for its functions during several developmental processes including cell fate specification, cell migration and axon guidance, and its members play fundamental roles in hippocampal development. The hippocampus is a structure that displays striking activity dependent plasticity. We examined whether LIM-HD genes and their co-factors are regulated during kainic acid induced seizure in the adult rat hippocampus as well as in early postnatal rats, when the hippocampal circuitry is not fully developed. We report a distinct and field-specific regulation of LIM-HD genes
Lhx1,Lhx2, and
Lhx9, LIM-only gene
Lmo4, and cofactor
Clim1a in the adult hippocampus after seizure induction. In contrast none of these genes displayed altered levels upon induction of seizure in postnatal animals. Our results provide evidence of temporal and spatial seizure mediated regulation of LIM-HD family members and suggest that LIM-HD gene function may be involved in activity dependent plasticity in the adult hippocampus
Collapse
Affiliation(s)
- Vanisha Lakhina
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Lewis Sigler Institute for Integrative Genomics, Princeton University, NJ, USA
| | - Lakshmi Subramanian
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Department of Neurology, University of California, San Francisco, CA, USA
| | - Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India ; Current affiliation: Cold Spring Harbor Laboratory, NY, USA
| | - Ashwin S Shetty
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
40
|
Wang ZI, Alexopoulos AV, Jones SE, Jaisani Z, Najm IM, Prayson RA. The pathology of magnetic-resonance-imaging-negative epilepsy. Mod Pathol 2013; 26:1051-8. [PMID: 23558575 DOI: 10.1038/modpathol.2013.52] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 11/09/2022]
Abstract
Patients with magnetic-resonance-imaging (MRI)-negative (or 'nonlesional') pharmacoresistant focal epilepsy are the most challenging group undergoing presurgical evaluation. Few large-scale studies have systematically reviewed the pathological substrates underlying MRI-negative epilepsies. In the current study, histopathological specimens were retrospectively reviewed from MRI-negative epilepsy patients (n=95, mean age=30 years, 50% female subjects). Focal cortical dysplasia cases were classified according to the International League Against Epilepsy (ILAE) and Palmini et al classifications. The most common pathologies found in this MRI-negative cohort included: focal cortical dysplasia (n=43, 45%), gliosis (n=21, 22%), hamartia+gliosis (n=12, 13%), and hippocampal sclerosis (n=9, 9%). The majority of focal cortical dysplasia were ILAE type I (n=37) or Palmini type I (n=39). Seven patients had no identifiable pathological abnormalities. The existence of positive pathology was not significantly associated with age or temporal/extratemporal resection. Follow-up data post surgery was available in 90 patients; 63 (70%) and 57 (63%) attained seizure freedom at 6 and 12 months, respectively. The finding of positive pathology was significantly associated with seizure-free outcome at 6 months (P=0.035), but not at 12 months. In subgroup analysis, the focal cortical dysplasia group was not significantly correlated with seizure-free outcome, as compared with the negative-pathology groups at either 6 or 12 months. Of note, the finding of hippocampal sclerosis had a significant positive correlation with seizure-free outcome when compared with the negative-pathology group (P=0.009 and 0.004 for 6- and 12-month outcome, respectively). Absence of a significant histopathology in the resected surgical specimen did not preclude seizure freedom. In conclusion, our study highlights the heterogeneity of epileptic pathologies in MRI-negative epilepsies, with focal cortical dysplasia being the most common finding. The existence of positive pathology in surgical specimen may be a good indication for short-term good seizure outcome. There is a small subset of cases in which no pathological abnormalities are identified.
Collapse
Affiliation(s)
- Z Irene Wang
- Cleveland Clinic Epilepsy Center, Cleveland, OH, USA
| | | | | | | | | | | |
Collapse
|
41
|
The influence of ectopic migration of granule cells into the hilus on dentate gyrus-CA3 function. PLoS One 2013; 8:e68208. [PMID: 23840835 PMCID: PMC3695928 DOI: 10.1371/journal.pone.0068208] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/27/2013] [Indexed: 11/29/2022] Open
Abstract
Postnatal neurogenesis of granule cells (GCs) in the dentate gyrus (DG) produces GCs that normally migrate from the subgranular zone to the GC layer. However, GCs can mismigrate into the hilus, the opposite direction. Previous descriptions of these hilar ectopic GCs (hEGCs) suggest that they are rare unless there are severe seizures. However, it is not clear if severe seizures are required, and it also is unclear if severe seizures are responsible for the abnormalities of hEGCs, which include atypical dendrites and electrophysiological properties. Here we show that large numbers of hEGCs develop in a transgenic mouse without severe seizures. The mice have a deletion of BAX, which normally regulates apoptosis. Surprisingly, we show that hEGCs in the BAX-/- mouse have similar abnormalities as hEGCs that arise after severe seizures. We next asked if there are selective effects of hEGCs, i.e., whether a robust population of hEGCs would have any effect on the DG if they were induced without severe seizures. Indeed, this appears to be true, because it has been reported that BAX-/- mice have defects in a behavior that tests pattern separation, which depends on the DG. However, inferring functional effects of hEGCs is difficult in mice with a constitutive BAX deletion because there is decreased apoptosis in and outside the DG. Therefore, a computational model of the normal DG and hippocampal subfield CA3 was used. Adding a small population of hEGCs (5% of all GCs), with characteristics defined empirically, was sufficient to disrupt a simulation of pattern separation and completion. Modeling results also showed that effects of hEGCs were due primarily to “backprojections” of CA3 pyramidal cell axons to the hilus. The results suggest that hEGCs can develop for diverse reasons, do not depend on severe seizures, and a small population of hEGCs may impair DG-dependent function.
Collapse
|
42
|
The endogenous regenerative capacity of the damaged newborn brain: boosting neurogenesis with mesenchymal stem cell treatment. J Cereb Blood Flow Metab 2013; 33:625-34. [PMID: 23403379 PMCID: PMC3652688 DOI: 10.1038/jcbfm.2013.3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neurogenesis continues throughout adulthood. The neurogenic capacity of the brain increases after injury by, e.g., hypoxia-ischemia. However, it is well known that in many cases brain damage does not resolve spontaneously, indicating that the endogenous regenerative capacity of the brain is insufficient. Neonatal encephalopathy leads to high mortality rates and long-term neurologic deficits in babies worldwide. Therefore, there is an urgent need to develop more efficient therapeutic strategies. The latest findings indicate that stem cells represent a novel therapeutic possibility to improve outcome in models of neonatal encephalopathy. Transplanted stem cells secrete factors that stimulate and maintain neurogenesis, thereby increasing cell proliferation, neuronal differentiation, and functional integration. Understanding the molecular and cellular mechanisms underlying neurogenesis after an insult is crucial for developing tools to enhance the neurogenic capacity of the brain. The aim of this review is to discuss the endogenous capacity of the neonatal brain to regenerate after a cerebral ischemic insult. We present an overview of the molecular and cellular mechanisms underlying endogenous regenerative processes during development as well as after a cerebral ischemic insult. Furthermore, we will consider the potential to use stem cell transplantation as a means to boost endogenous neurogenesis and restore brain function.
Collapse
|
43
|
Jafari M, Haist V, Baumgärtner W, Wagner S, Stein VM, Tipold A, Wendt H, Potschka H. Impact of Theiler's virus infection on hippocampal neuronal progenitor cells: differential effects in two mouse strains. Neuropathol Appl Neurobiol 2013; 38:647-64. [PMID: 22288387 DOI: 10.1111/j.1365-2990.2012.01256.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Disease-associated alterations in hippocampal neurogenesis are discussed as an important factor contributing to long-term consequences of central nervous system diseases. Therefore, the study aimed to determine the impact of Theiler's murine encephalomyelitis virus infection on hippocampal cell proliferation, neuronal progenitor cells and neurogenesis as well as the influence of microglia on respective disease-associated alterations. METHODS The impact of the infection was evaluated in two mouse strains which differ in the disease course, with an acute polioencephalitis followed by virus elimination in C57BL/6 mice and a chronic demyelinating disease in SJL/J mice. RESULTS Infection with the low neurovirulent BeAn strain did not exert significant acute effects regardless of the mouse strain. In the chronic phase, the number of neuronal progenitor cells and early postmitotic neurones was significantly reduced in infected SJL/J mice, whereas no long-term alterations were observed in C57BL/6 mice. A contrasting course of microglia activation was observed in the two mouse strains, with an early increase in the number of activated microglia cells in SJL/J mice and a delayed increase in C57BL/6 mice. Quantitative analysis did not confirm a correlation between the number of activated microglia and the number of neuronal progenitor cells and early postmitotic neurones. However, flow cytometric analyses revealed alterations in the functional state of microglial cells which might have affected the generation of neuronal progenitor cells. CONCLUSIONS Theiler's murine encephalomyelitis virus infection can exert delayed effects on the hippocampal neuronal progenitor population with long-term alterations evident 3 months following infection. These alterations proved to depend on strain susceptibility and might contribute to detrimental consequences of virus encephalitis such as cognitive impairment.
Collapse
Affiliation(s)
- M Jafari
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Licko T, Seeger N, Zellinger C, Russmann V, Matagne A, Potschka H. Lacosamide treatment following status epilepticus attenuates neuronal cell loss and alterations in hippocampal neurogenesis in a rat electrical status epilepticus model. Epilepsia 2013; 54:1176-85. [PMID: 23614482 DOI: 10.1111/epi.12196] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 12/22/2022]
Abstract
PURPOSE The antiepileptic drug, lacosamide, exerts its therapeutic activity by enhancing slow inactivation of voltage-gated sodium channels. Because putative preventive or disease-modifying effects of drugs may affect epileptogenesis, intrinsic severity, and comorbidities, it is of particular interest to assess the effect of lacosamide on the development of epilepsy and associated cellular alterations. METHODS The effect of lacosamide was evaluated in an electrical rat status epilepticus (SE) model with a 24-day treatment phase following induction of SE. The impact of lacosamide on the development of spontaneous seizures based on continuous video-electroencephalography (EEG) monitoring, as well as the impact on neuronal cell loss and alterations in hippocampal neurogenesis, was assessed. KEY FINDINGS Neither low-dose nor high-dose lacosamide affected the development of spontaneous seizures. A dose-dependent neuroprotective effect of lacosamide with significant reduction of neuronal cell loss was observed in the hippocampal CA1 region, as well as in the piriform cortex. In addition, lacosamide attenuated the impact of SE on the rate of hippocampal cell neurogenesis. Moreover, lacosamide prevented a significant rise in the number of persistent basal dendrites. SIGNIFICANCE Our data do not support an antiepileptogenic effect of lacosamide. However, because lacosamide reduced SE-associated cellular alterations, it would be of interest to determine whether these effects indicate a putative disease-modifying effect of lacosamide in future studies.
Collapse
Affiliation(s)
- Thomas Licko
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilian-University, Munich, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Chin J, Scharfman HE. Shared cognitive and behavioral impairments in epilepsy and Alzheimer's disease and potential underlying mechanisms. Epilepsy Behav 2013; 26:343-51. [PMID: 23321057 PMCID: PMC3924321 DOI: 10.1016/j.yebeh.2012.11.040] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 11/17/2012] [Indexed: 01/27/2023]
Abstract
Seizures in patients with Alzheimer's disease (AD) have been examined by many investigators over the last several decades, and there are diverse opinions about their potential relevance to AD pathophysiology. Some studies suggest that seizures appear to be a fairly uncommon co-morbidity, whereas other studies report a higher incidence of seizures in patients with AD. It was previously thought that seizures play a minor role in AD pathophysiology because of their low frequency, and also because they may only be noticed during late stages of AD, suggesting that seizures are likely to be a consequence of neurodegeneration rather than a contributing factor. However, clinical reports indicate that seizures can occur early in the emergence of AD symptoms, particularly in familial AD. In this case, seizures may be an integral part of the emerging pathophysiology. This view has been supported by evidence of recurrent spontaneous seizures in transgenic mouse models of AD in which familial AD is simulated. Additional data from transgenic animals suggest that there may be a much closer relationship between seizures and AD than previously considered. There is also evidence that seizures facilitate production of amyloid β (Aβ) and can cause impairments in cognition and behavior in both animals and humans. However, whether seizures play a role in the early stages of AD pathogenesis is still debated. Therefore, it is timely to review the similarities and differences between AD and epilepsy, as well as data suggesting that seizures may contribute to cognitive and behavioral dysfunction in AD. Here we focus on AD and temporal lobe epilepsy (TLE), a particular type of epilepsy that involves the temporal lobe, a region that influences behavior and is critical to memory. We also consider potential neurobiological mechanisms that support the view that the causes of seizures in TLE may be related to the causes of cognitive dysfunction in AD. We suggest that similar underlying mechanisms may exist for at least some of the aspects of AD that are also found in TLE.
Collapse
Affiliation(s)
- Jeannie Chin
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Helen E. Scharfman
- Child & Adolescent Psychiatry, Physiology & Neuroscience, Psychiatry, New York University Langone Medical Center, New York, NY, USA,Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA,Correspondence to: H.E. Scharfman, Child & Adolescent Psychiatry, Physiology & Neuroscience, Psychiatry, New York University Langone Medical Center, New York, NY, USA. (H.E. Scharfman)
| |
Collapse
|
46
|
Neurogenesis in temporal lobe epilepsy: Relationship between histological findings and changes in dentate gyrus proliferative properties. Clin Neurol Neurosurg 2013; 115:187-91. [DOI: 10.1016/j.clineuro.2012.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Revised: 04/02/2012] [Accepted: 05/12/2012] [Indexed: 11/24/2022]
|
47
|
Choi YS, Karelina K, Alzate-Correa D, Hoyt KR, Impey S, Arthur JS, Obrietan K. Mitogen- and stress-activated kinases regulate progenitor cell proliferation and neuron development in the adult dentate gyrus. J Neurochem 2012; 123:676-88. [PMID: 23020821 PMCID: PMC3575744 DOI: 10.1111/jnc.12035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/21/2012] [Accepted: 09/25/2012] [Indexed: 01/19/2023]
Abstract
The neurogenic niche within the subgranular zone (SGZ) of the dentate gyrus is a source of new neurons throughout life. Interestingly, SGZ proliferative capacity is regulated by both physiological and pathophysiological conditions. One outstanding question involves the molecular mechanisms that regulate both basal and inducible adult neurogenesis. Here, we examined the role of the MAPK-regulated kinases, mitogen- and stress-activated kinase (MSK)1 and MSK2. as regulators of dentate gyrus SGZ progenitor cell proliferation and neurogenesis. Under basal conditions, MSK1/2 null mice exhibited significantly reduced progenitor cell proliferation capacity and a corollary reduction in the number of doublecortin (DCX)-positive immature neurons. Strikingly, seizure-induced progenitor proliferation was totally blocked in MSK1/2 null mice. This blunting of cell proliferation in MSK1/2 null mice was partially reversed by forskolin infusion, indicating that the inducible proliferative capacity of the progenitor cell population was intact. Furthermore, in MSK1/2 null mice, DCX-positive immature neurons exhibited reduced neurite arborization. Together, these data reveal a critical role for MSK1/2 as regulators of both basal and activity-dependent progenitor cell proliferation and morphological maturation in the SGZ.
Collapse
Affiliation(s)
- Yun-Sik Choi
- Department of Pharmaceutical Science & Technology, Catholic University of Daegu, Gyeongbuk, Rep. of Korea
| | - Kate Karelina
- Department of Neuroscience, Ohio State University, Columbus, OH 43210
| | | | - Kari R. Hoyt
- Division of Pharmacology, Ohio State University, Columbus, OH 43210
| | - Soren Impey
- Department of Cell and Developmental Biology, Oregon Health & Sciences University, Portland, OR 97239
| | - J. Simon Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH 43210
| |
Collapse
|
48
|
Jafari M, Soerensen J, Bogdanović RM, Dimou L, Götz M, Potschka H. Long-term genetic fate mapping of adult generated neurons in a mouse temporal lobe epilepsy model. Neurobiol Dis 2012; 48:454-63. [DOI: 10.1016/j.nbd.2012.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 06/15/2012] [Accepted: 06/22/2012] [Indexed: 11/25/2022] Open
|
49
|
von Rüden EL, Avemary J, Zellinger C, Algermissen D, Bock P, Beineke A, Baumgärtner W, Stein VM, Tipold A, Potschka H. Distemper virus encephalitis exerts detrimental effects on hippocampal neurogenesis. Neuropathol Appl Neurobiol 2012; 38:426-42. [PMID: 21883377 DOI: 10.1111/j.1365-2990.2011.01218.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS Despite knowledge about the impact of brain inflammation on hippocampal neurogenesis, data on the influence of virus encephalitis on dentate granule cell neurogenesis are so far limited. Canine distemper is considered an interesting model of virus encephalitis, which can be associated with a chronic progressing disease course and can cause symptomatic seizures. METHODS To determine the impact of canine distemper virus (CDV) infection on hippocampal neurogenesis, we compared post-mortem tissue from dogs with infection with and without seizures, from epileptic dogs with non-viral aetiology and from dogs without central nervous system diseases. RESULTS The majority of animals with infection and with epilepsy of non-viral aetiology exhibited neuronal progenitor numbers below the age average in controls. Virus infection with and without seizures significantly decreased the mean number of neuronal progenitor cells by 43% and 76% as compared to age-matched controls. Ki-67 labelling demonstrated that hippocampal cell proliferation was neither affected by infection nor by epilepsy of non-viral aetiology. Analysis of CDV infection in cells expressing caspase-3, doublecortin or Ki-67 indicated that infection of neuronal progenitor cells is extremely rare and suggests that infection might damage non-differentiated progenitor cells, hamper neuronal differentiation and promote glial differentiation. A high inter-individual variance in the number of lectin-reactive microglial cells was evident in dogs with distemper infection. Statistical analyses did not reveal a correlation between the number of lectin-reactive microglia cells and neuronal progenitor cells. CONCLUSIONS Our data demonstrate that virus encephalitis with and without seizures can exert detrimental effects on hippocampal neurogenesis, which might contribute to long-term consequences of the disease. The lack of a significant impact of distemper virus on Ki-67-labelled cells indicates that the infection affected neuronal differentiation and survival of newborn cells rather than hippocampal cell proliferation.
Collapse
Affiliation(s)
- E-L von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kobow K, Auvin S, Jensen F, Löscher W, Mody I, Potschka H, Prince D, Sierra A, Simonato M, Pitkänen A, Nehlig A, Rho JM. Finding a better drug for epilepsy: antiepileptogenesis targets. Epilepsia 2012; 53:1868-76. [PMID: 23061663 DOI: 10.1111/j.1528-1167.2012.03716.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
For several decades, both in vitro and in vivo models of seizures and epilepsy have been employed to unravel the molecular and cellular mechanisms underlying the occurrence of spontaneous recurrent seizures (SRS)-the defining hallmark of the epileptic brain. However, despite great advances in our understanding of seizure genesis, investigators have yet to develop reliable biomarkers and surrogate markers of the epileptogenic process. Sadly, the pathogenic mechanisms that produce the epileptic condition, especially after precipitating events such as head trauma, inflammation, or prolonged febrile convulsions, are poorly understood. A major challenge has been the inherent complexity and heterogeneity of known epileptic syndromes and the differential genetic susceptibilities exhibited by patients at risk. Therefore, it is unlikely that there is only one fundamental pathophysiologic mechanism shared by all the epilepsies. Identification of antiepileptogenesis targets has been an overarching goal over the last decade, as current anticonvulsant medications appear to influence only the acute process of ictogenesis. Clearly, there is an urgent need to develop novel therapeutic interventions that are disease modifying-therapies that either completely or partially prevent the emergence of SRS. An important secondary goal is to develop new treatments that can also lessen the burden of epilepsy comorbidities (e.g., cognitive impairment, mood disorders) by preventing or reducing the deleterious changes during the epileptogenic process. This review summarizes novel antiepileptogenesis targets that were critically discussed at the XIth Workshop on the Neurobiology of Epilepsy (WONOEP XI) meeting in Grottaferrata, Italy. Further, emerging neurometabolic links among several target mechanisms and highlights of the panel discussion are presented.
Collapse
Affiliation(s)
- Katja Kobow
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|